Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Cell ; 184(3): 810-826.e23, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33406409

ABSTRACT

Development of the human intestine is not well understood. Here, we link single-cell RNA sequencing and spatial transcriptomics to characterize intestinal morphogenesis through time. We identify 101 cell states including epithelial and mesenchymal progenitor populations and programs linked to key morphogenetic milestones. We describe principles of crypt-villus axis formation; neural, vascular, mesenchymal morphogenesis, and immune population of the developing gut. We identify the differentiation hierarchies of developing fibroblast and myofibroblast subtypes and describe diverse functions for these including as vascular niche cells. We pinpoint the origins of Peyer's patches and gut-associated lymphoid tissue (GALT) and describe location-specific immune programs. We use our resource to present an unbiased analysis of morphogen gradients that direct sequential waves of cellular differentiation and define cells and locations linked to rare developmental intestinal disorders. We compile a publicly available online resource, spatio-temporal analysis resource of fetal intestinal development (STAR-FINDer), to facilitate further work.


Subject(s)
Intestines/cytology , Intestines/growth & development , Single-Cell Analysis , Endothelial Cells/cytology , Enteric Nervous System/cytology , Fetus/embryology , Fibroblasts/cytology , Humans , Immunity , Intestinal Diseases/congenital , Intestinal Diseases/pathology , Intestinal Mucosa/growth & development , Intestines/blood supply , Ligands , Mesoderm/cytology , Neovascularization, Physiologic , Pericytes/cytology , Stem Cells/cytology , Time Factors , Transcription Factors/metabolism
2.
Nat Immunol ; 23(1): 50-61, 2022 01.
Article in English | MEDLINE | ID: mdl-34853448

ABSTRACT

NP105-113-B*07:02-specific CD8+ T cell responses are considered among the most dominant in SARS-CoV-2-infected individuals. We found strong association of this response with mild disease. Analysis of NP105-113-B*07:02-specific T cell clones and single-cell sequencing were performed concurrently, with functional avidity and antiviral efficacy assessed using an in vitro SARS-CoV-2 infection system, and were correlated with T cell receptor usage, transcriptome signature and disease severity (acute n = 77, convalescent n = 52). We demonstrated a beneficial association of NP105-113-B*07:02-specific T cells in COVID-19 disease progression, linked with expansion of T cell precursors, high functional avidity and antiviral effector function. Broad immune memory pools were narrowed postinfection but NP105-113-B*07:02-specific T cells were maintained 6 months after infection with preserved antiviral efficacy to the SARS-CoV-2 Victoria strain, as well as Alpha, Beta, Gamma and Delta variants. Our data show that NP105-113-B*07:02-specific T cell responses associate with mild disease and high antiviral efficacy, pointing to inclusion for future vaccine design.


Subject(s)
HLA-B7 Antigen/immunology , Immunodominant Epitopes/immunology , Nucleocapsid Proteins/immunology , SARS-CoV-2/immunology , T-Lymphocytes, Cytotoxic/immunology , Aged , Amino Acid Sequence , Antibodies, Viral/immunology , Antibody Affinity/immunology , COVID-19/immunology , COVID-19/pathology , Cell Line, Transformed , Female , Gene Expression Profiling , Humans , Immunologic Memory/immunology , Male , Middle Aged , Receptors, Antigen, T-Cell/immunology , Severity of Illness Index , Vaccinia virus/genetics , Vaccinia virus/immunology , Vaccinia virus/metabolism
3.
Cell ; 175(2): 372-386.e17, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30270042

ABSTRACT

Intestinal mesenchymal cells play essential roles in epithelial homeostasis, matrix remodeling, immunity, and inflammation. But the extent of heterogeneity within the colonic mesenchyme in these processes remains unknown. Using unbiased single-cell profiling of over 16,500 colonic mesenchymal cells, we reveal four subsets of fibroblasts expressing divergent transcriptional regulators and functional pathways, in addition to pericytes and myofibroblasts. We identified a niche population located in proximity to epithelial crypts expressing SOX6, F3 (CD142), and WNT genes essential for colonic epithelial stem cell function. In colitis, we observed dysregulation of this niche and emergence of an activated mesenchymal population. This subset expressed TNF superfamily member 14 (TNFSF14), fibroblastic reticular cell-associated genes, IL-33, and Lysyl oxidases. Further, it induced factors that impaired epithelial proliferation and maturation and contributed to oxidative stress and disease severity in vivo. Our work defines how the colonic mesenchyme remodels to fuel inflammation and barrier dysfunction in IBD.


Subject(s)
Inflammatory Bowel Diseases/physiopathology , Mesoderm/physiology , Animals , Cell Proliferation , Colitis/genetics , Colitis/physiopathology , Colon/physiology , Epithelial Cells/metabolism , Fibroblasts/physiology , Genetic Heterogeneity , Homeostasis , Humans , Inflammation , Intestinal Mucosa/immunology , Intestinal Mucosa/physiology , Intestines/immunology , Intestines/physiology , Mesenchymal Stem Cells/physiology , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Myofibroblasts , Pericytes , RAW 264.7 Cells , SOXD Transcription Factors/physiology , Single-Cell Analysis/methods , Thromboplastin/physiology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Wnt Signaling Pathway/physiology
4.
Nature ; 587(7834): 460-465, 2020 11.
Article in English | MEDLINE | ID: mdl-33149301

ABSTRACT

Atrial fibrillation, the most common cardiac arrhythmia, is an important contributor to mortality and morbidity, and particularly to the risk of stroke in humans1. Atrial-tissue fibrosis is a central pathophysiological feature of atrial fibrillation that also hampers its treatment; the underlying molecular mechanisms are poorly understood and warrant investigation given the inadequacy of present therapies2. Here we show that calcitonin, a hormone product of the thyroid gland involved in bone metabolism3, is also produced by atrial cardiomyocytes in substantial quantities and acts as a paracrine signal that affects neighbouring collagen-producing fibroblasts to control their proliferation and secretion of extracellular matrix proteins. Global disruption of calcitonin receptor signalling in mice causes atrial fibrosis and increases susceptibility to atrial fibrillation. In mice in which liver kinase B1 is knocked down specifically in the atria, atrial-specific knockdown of calcitonin promotes atrial fibrosis and increases and prolongs spontaneous episodes of atrial fibrillation, whereas atrial-specific overexpression of calcitonin prevents both atrial fibrosis and fibrillation. Human patients with persistent atrial fibrillation show sixfold lower levels of myocardial calcitonin compared to control individuals with normal heart rhythm, with loss of calcitonin receptors in the fibroblast membrane. Although transcriptome analysis of human atrial fibroblasts reveals little change after exposure to calcitonin, proteomic analysis shows extensive alterations in extracellular matrix proteins and pathways related to fibrogenesis, infection and immune responses, and transcriptional regulation. Strategies to restore disrupted myocardial calcitonin signalling thus may offer therapeutic avenues for patients with atrial fibrillation.


Subject(s)
Arrhythmias, Cardiac/metabolism , Calcitonin/metabolism , Fibrinogen/biosynthesis , Heart Atria/metabolism , Myocardium/metabolism , Paracrine Communication , Animals , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Atrial Fibrillation , Collagen Type I/metabolism , Female , Fibroblasts/metabolism , Fibrosis/metabolism , Fibrosis/pathology , Heart Atria/cytology , Heart Atria/pathology , Heart Atria/physiopathology , Humans , Male , Mice , Myocardium/cytology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Receptors, Calcitonin/metabolism
5.
Nature ; 567(7746): 49-55, 2019 03.
Article in English | MEDLINE | ID: mdl-30814735

ABSTRACT

The colonic epithelium facilitates host-microorganism interactions to control mucosal immunity, coordinate nutrient recycling and form a mucus barrier. Breakdown of the epithelial barrier underpins inflammatory bowel disease (IBD). However, the specific contributions of each epithelial-cell subtype to this process are unknown. Here we profile single colonic epithelial cells from patients with IBD and unaffected controls. We identify previously unknown cellular subtypes, including gradients of progenitor cells, colonocytes and goblet cells within intestinal crypts. At the top of the crypts, we find a previously unknown absorptive cell, expressing the proton channel OTOP2 and the satiety peptide uroguanylin, that senses pH and is dysregulated in inflammation and cancer. In IBD, we observe a positional remodelling of goblet cells that coincides with downregulation of WFDC2-an antiprotease molecule that we find to be expressed by goblet cells and that inhibits bacterial growth. In vivo, WFDC2 preserves the integrity of tight junctions between epithelial cells and prevents invasion by commensal bacteria and mucosal inflammation. We delineate markers and transcriptional states, identify a colonic epithelial cell and uncover fundamental determinants of barrier breakdown in IBD.


Subject(s)
Colon/cytology , Colon/pathology , Epithelial Cells/classification , Epithelial Cells/cytology , Health , Inflammatory Bowel Diseases/pathology , Ion Channels/metabolism , Animals , Biomarkers/analysis , Colitis, Ulcerative/genetics , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Colon/microbiology , Epithelial Cells/microbiology , Epithelial Cells/pathology , Genetic Predisposition to Disease/genetics , Goblet Cells/cytology , Goblet Cells/metabolism , Goblet Cells/pathology , Humans , Hydrogen-Ion Concentration , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Natriuretic Peptides/metabolism , Proteins/metabolism , Single-Cell Analysis , Stem Cells/cytology , Stem Cells/metabolism , Stem Cells/pathology , Tight Junctions/metabolism , Transcription, Genetic , WAP Four-Disulfide Core Domain Protein 2
6.
Proc Natl Acad Sci U S A ; 115(44): 11192-11197, 2018 10 30.
Article in English | MEDLINE | ID: mdl-30322920

ABSTRACT

To elucidate cellular diversity and clonal evolution in tissues and tumors, one must resolve genomic heterogeneity in single cells. To this end, we have developed low-cost, mass-producible micro-/nanofluidic chips for DNA extraction from individual cells. These chips have modules that collect genomic DNA for sequencing or map genomic structure directly, on-chip, with denaturation-renaturation (D-R) optical mapping [Marie R, et al. (2013) Proc Natl Acad Sci USA 110:4893-4898]. Processing of single cells from the LS174T colorectal cancer cell line showed that D-R mapping of single molecules can reveal structural variation (SV) in the genome of single cells. In one experiment, we processed 17 fragments covering 19.8 Mb of the cell's genome. One megabase-large fragment aligned well to chromosome 19 with half its length, while the other half showed variable alignment. Paired-end single-cell sequencing supported this finding, revealing a region of complexity and a 50-kb deletion. Sequencing struggled, however, to detect a 20-kb gap that D-R mapping showed clearly in a megabase fragment that otherwise mapped well to the reference at the pericentromeric region of chromosome 4. Pericentromeric regions are complex and show substantial sequence homology between different chromosomes, making mapping of sequence reads ambiguous. Thus, D-R mapping directly, from a single molecule, revealed characteristics of the single-cell genome that were challenging for short-read sequencing.


Subject(s)
Chromosome Mapping/methods , DNA/genetics , Genome/genetics , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, DNA/methods , Cell Line, Tumor , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 4/genetics , Clonal Evolution/genetics , Colorectal Neoplasms/genetics , Genomics/methods , Humans , Sequence Deletion/genetics
7.
J Pathol ; 247(3): 293-304, 2019 03.
Article in English | MEDLINE | ID: mdl-30306567

ABSTRACT

Colonic epithelial cells are highly polarised with a lumen-facing apical membrane, termed the brush border, and a basal membrane in contact with the underlying extracellular matrix (ECM). This polarity is often maintained in cancer tissue in the form of neoplastic glands and has prognostic value. We compared the cellular polarity of several ex vivo spheroid colonic cancer cultures with their parental tumours and found that those grown as non-attached colonies exhibited apical brush border proteins on their outer cellular membranes. Transfer of these cultures to an ECM, such as collagen, re-established the centralised apical polarity observed in vivo. The multidrug resistance protein ABCB1 also became aberrantly polarised to outer colony membranes in suspension cultures, unlike cultures grown in collagen, where it was polarised to central lumens. This polarity switch was dependent on the presence of serum or selected serum components, including epidermal growth factor (EGF), transforming growth factor-ß1 (TGF-ß1) and insulin-like growth factor-1 (IGF-1). The apical/basal orientation of primary cancer colon cultures cultured in collagen/serum was modulated by α2ß1 integrin signalling. The polarisation of ABCB1 in colonies significantly altered drug uptake and sensitivity, as the outward polarisation of ABCB1 in suspension colonies effluxed substrates more effectively than ECM-grown colonies with ABCB1 polarised to central lumens. Thus, serum-free suspension colonies were more resistant to a variety of anti-cancer drugs than ECM-grown colonies. In conclusion, the local stroma, or absence thereof, can have profound effects on the sensitivity of colorectal cultures to drugs that are ABCB1 substrates. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Cell Polarity/physiology , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/physiology , ATP Binding Cassette Transporter, Subfamily B/physiology , Actins/metabolism , Collagen/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Drug Combinations , Extracellular Matrix/metabolism , Humans , Integrin beta1/physiology , Laminin , Proteoglycans , Signal Transduction/physiology , Spheroids, Cellular/pathology , Tumor Cells, Cultured
8.
Proc Natl Acad Sci U S A ; 113(15): E2162-71, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27036009

ABSTRACT

Pericryptal myofibroblasts in the colon and rectum play an important role in regulating the normal colorectal stem cell niche and facilitating tumor progression. Myofibroblasts previously have been distinguished from normal fibroblasts mostly by the expression of α smooth muscle actin (αSMA). We now have identified AOC3 (amine oxidase, copper containing 3), a surface monoamine oxidase, as a new marker of myofibroblasts by showing that it is the target protein of the myofibroblast-reacting mAb PR2D3. The normal and tumor tissue distribution and the cell line reactivity of AOC3 match that expected for myofibroblasts. We have shown that the surface expression of AOC3 is sensitive to digestion by trypsin and collagenase and that anti-AOC3 antibodies can be used for FACS sorting of myofibroblasts obtained by nonenzymatic procedures. Whole-genome microarray mRNA-expression profiles of myofibroblasts and skin fibroblasts revealed four additional genes that are significantly differentially expressed in these two cell types: NKX2-3 and LRRC17 in myofibroblasts and SHOX2 and TBX5 in skin fibroblasts. TGFß substantially down-regulated AOC3 expression in myofibroblasts but in skin fibroblasts it dramatically increased the expression of αSMA. A knockdown of NKX2-3 in myofibroblasts caused a decrease of myofibroblast-related gene expression and increased expression of the fibroblast-associated gene SHOX2, suggesting that NKX2-3 is a key mediator for maintaining myofibroblast characteristics. Our results show that colorectal myofibroblasts, as defined by the expression of AOC3, NKX2-3, and other markers, are a distinctly different cell type from TGFß-activated fibroblasts.


Subject(s)
Amine Oxidase (Copper-Containing)/metabolism , Cell Adhesion Molecules/metabolism , Fibroblasts/metabolism , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Amine Oxidase (Copper-Containing)/genetics , Cell Adhesion Molecules/genetics , Cell Differentiation/genetics , Cell Line , Cell Line, Tumor , Cells, Cultured , Colon/cytology , Colon/metabolism , Colorectal Neoplasms/metabolism , Fibroblasts/cytology , Gene Expression , Homeodomain Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins , Oligonucleotide Array Sequence Analysis , Proteins/genetics , RNA, Messenger/metabolism , Skin/cytology , Skin/metabolism , T-Box Domain Proteins/genetics , Transcription Factors/genetics , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured
10.
J Pathol ; 234(1): 34-45, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24797403

ABSTRACT

We have developed a simple procedure for deriving pure cultures of growing cancer cells from colorectal cancers, including material refrigerated overnight, for pathological characterization and cytotoxicity assays. Forty-six cancers were processed and cultures set up under varying culture conditions. Use of a Rho kinase (ROCK1) inhibitor markedly increased culture survival, resulting in 80% of samples growing in culture for at least 1 month and beyond. Overnight refrigeration of samples before culture initiation had little effect on success rates, paving the way for cultures to be established for samples collected over wide geographical areas, such as those for clinical trials. Primary cultures demonstrated good correlation for differentiation markers compared to parent cancers, and were highly dynamic in 3D culture. In Matrigel, many colonies formed central lumens, indicating the presence of stem-like cells. Viable colonies in these cultures recapitulated the in vivo generation of carcinoembryonic antigen (CEA)-positive necrotic/apoptotic debris, much of which was derived from abnormal vacuolated dynamic 'bubble cells' that have not previously been described. Although bubble cells morphologically resembled signet ring cells, a rare cancer subtype, immunostaining suggested that they were most likely derived from terminally differentiated enterocytes. Micro-assays showed that drug toxicity could be measured in these cultures within hours and with sensitivity down to a few hundred cells. Primary cultures derived by our method provide valid in vitro avatars for studying the pathology of cancers in vitro and are amenable to pre-clinical drug testing, paving the way for personalized cancer treatment.


Subject(s)
Adenocarcinoma/pathology , Colorectal Neoplasms/pathology , Goblet Cells/pathology , Spheroids, Cellular/pathology , Cell Differentiation , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Goblet Cells/drug effects , Humans , Keratin-20/metabolism , Spheroids, Cellular/drug effects , Staurosporine/pharmacology , Time Factors , Tumor Cells, Cultured
11.
BMC Genomics ; 15: 487, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24942464

ABSTRACT

Understanding the links between genetic, epigenetic and non-genetic factors throughout the lifespan and across generations and their role in disease susceptibility and disease progression offer entirely new avenues and solutions to major problems in our society. To overcome the numerous challenges, we have come up with nine major conclusions to set the vision for future policies and research agendas at the European level.


Subject(s)
Epigenesis, Genetic , Genome , Research , Epigenomics , Genomics , Humans
12.
Cancer Res ; 84(2): 211-225, 2024 01 16.
Article in English | MEDLINE | ID: mdl-37921711

ABSTRACT

Myelodysplastic syndromes with ring sideroblasts (MDS-RS) commonly develop from hematopoietic stem cells (HSC) bearing mutations in the splicing factor SF3B1 (SF3B1mt). Direct studies into MDS-RS pathobiology have been limited by a lack of model systems that fully recapitulate erythroid biology and RS development and the inability to isolate viable human RS. Here, we combined successful direct RS isolation from patient samples, high-throughput multiomics analysis of cells encompassing the SF3B1mt stem-erythroid continuum, and functional assays to investigate the impact of SF3B1mt on erythropoiesis and RS accumulation. The isolated RS differentiated, egressed into the blood, escaped traditional nonsense-mediated decay (NMD) mechanisms, and leveraged stress-survival pathways that hinder wild-type hematopoiesis through pathogenic GDF15 overexpression. Importantly, RS constituted a contaminant of magnetically enriched CD34+ cells, skewing bulk transcriptomic data. Mis-splicing in SF3B1mt cells was intensified by erythroid differentiation through accelerated RNA splicing and decreased NMD activity, and SF3B1mt led to truncations in several MDS-implicated genes. Finally, RNA mis-splicing induced an uncoupling of RNA and protein expression, leading to critical abnormalities in proapoptotic p53 pathway genes. Overall, this characterization of erythropoiesis in SF3B1mt RS provides a resource for studying MDS-RS and uncovers insights into the unexpectedly active biology of the "dead-end" RS. SIGNIFICANCE: Ring sideroblast isolation combined with state-of-the-art multiomics identifies survival mechanisms underlying SF3B1-mutant erythropoiesis and establishes an active role for erythroid differentiation and ring sideroblasts themselves in SF3B1-mutant myelodysplastic syndrome pathogenesis.


Subject(s)
Myelodysplastic Syndromes , Phosphoproteins , Humans , Phosphoproteins/genetics , Phosphoproteins/metabolism , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , RNA Splicing/genetics , Mutation , Transcription Factors/metabolism , RNA/metabolism
13.
Leukemia ; 37(2): 276-287, 2023 02.
Article in English | MEDLINE | ID: mdl-36572750

ABSTRACT

Nuclear factor I-C (NFIC) belongs to a family of NFI transcription factors that binds to DNA through CAATT-boxes and are involved in cellular differentiation and stem cell maintenance. Here we show NFIC protein is significantly overexpressed in 69% of acute myeloid leukemia patients. Examination of the functional consequences of NFIC overexpression in HSPCs showed that this protein promoted monocytic differentiation. Single-cell RNA sequencing analysis further demonstrated that NFIC overexpressing monocytes had increased expression of growth and survival genes. In contrast, depletion of NFIC through shRNA decreased cell growth, increased cell cycle arrest and apoptosis in AML cell lines and AML patient blasts. Further, in AML cell lines (THP-1), bulk RNA sequencing of NFIC knockdown led to downregulation of genes involved in cell survival and oncogenic signaling pathways including mixed lineage leukemia-1 (MLL-1). Lastly, we show that NFIC knockdown in an ex vivo mouse MLL::AF9 pre-leukemic stem cell model, decreased their growth and colony formation and increased expression of myeloid differentiation markers Gr1 and Mac1. Collectively, our results suggest that NFIC is an important transcription factor in myeloid differentiation as well as AML cell survival and is a potential therapeutic target in AML.


Subject(s)
Leukemia, Myeloid, Acute , NFI Transcription Factors , Animals , Mice , Cell Differentiation/physiology , Cell Survival/genetics , Hematopoiesis , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , NFI Transcription Factors/metabolism
14.
Nat Commun ; 12(1): 4797, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34376651

ABSTRACT

Sutures separate the flat bones of the skull and enable coordinated growth of the brain and overlying cranium. The coronal suture is most commonly fused in monogenic craniosynostosis, yet the unique aspects of its development remain incompletely understood. To uncover the cellular diversity within the murine embryonic coronal suture, we generated single-cell transcriptomes and performed extensive expression validation. We find distinct pre-osteoblast signatures between the bone fronts and periosteum, a ligament-like population above the suture that persists into adulthood, and a chondrogenic-like population in the dura mater underlying the suture. Lineage tracing reveals an embryonic Six2+ osteoprogenitor population that contributes to the postnatal suture mesenchyme, with these progenitors being preferentially affected in a Twist1+/-; Tcf12+/- mouse model of Saethre-Chotzen Syndrome. This single-cell atlas provides a resource for understanding the development of the coronal suture and the mechanisms for its loss in craniosynostosis.


Subject(s)
Cranial Sutures/metabolism , Gene Expression Regulation, Developmental , Osteogenesis/genetics , Single-Cell Analysis/methods , Transcriptome/genetics , Acrocephalosyndactylia/embryology , Acrocephalosyndactylia/genetics , Acrocephalosyndactylia/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cranial Sutures/cytology , Cranial Sutures/embryology , Dura Mater/cytology , Dura Mater/embryology , Dura Mater/metabolism , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Mice, Knockout , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/metabolism , RNA-Seq/methods , Skull/cytology , Skull/embryology , Skull/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
15.
J Exp Med ; 218(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33416891

ABSTRACT

Juvenile myelomonocytic leukemia (JMML) is a poor-prognosis childhood leukemia usually caused by RAS-pathway mutations. The cellular hierarchy in JMML is poorly characterized, including the identity of leukemia stem cells (LSCs). FACS and single-cell RNA sequencing reveal marked heterogeneity of JMML hematopoietic stem/progenitor cells (HSPCs), including an aberrant Lin-CD34+CD38-CD90+CD45RA+ population. Single-cell HSPC index-sorting and clonogenic assays show that (1) all somatic mutations can be backtracked to the phenotypic HSC compartment, with RAS-pathway mutations as a "first hit," (2) mutations are acquired with both linear and branching patterns of clonal evolution, and (3) mutant HSPCs are present after allogeneic HSC transplant before molecular/clinical evidence of relapse. Stem cell assays reveal interpatient heterogeneity of JMML LSCs, which are present in, but not confined to, the phenotypic HSC compartment. RNA sequencing of JMML LSC reveals up-regulation of stem cell and fetal genes (HLF, MEIS1, CNN3, VNN2, and HMGA2) and candidate therapeutic targets/biomarkers (MTOR, SLC2A1, and CD96), paving the way for LSC-directed disease monitoring and therapy in this disease.


Subject(s)
Hematopoietic Stem Cells/pathology , Leukemia, Myelomonocytic, Juvenile/pathology , Animals , Biomarkers, Tumor/genetics , Cell Line , Female , Humans , Leukemia, Myelomonocytic, Juvenile/genetics , Male , Mice , Mutation/genetics , Neoplastic Stem Cells/pathology , Signal Transduction/genetics , Up-Regulation/genetics
16.
Front Immunol ; 12: 623430, 2021.
Article in English | MEDLINE | ID: mdl-33746960

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is the most severe form of chronic lung fibrosis. Circulating monocytes have been implicated in immune pathology in IPF but their phenotype is unknown. In this work, we determined the immune phenotype of monocytes in IPF using multi-colour flow cytometry, RNA sequencing and corresponding serum factors, and mapped the main findings to amount of lung fibrosis and single cell transcriptomic landscape of myeloid cells in IPF lungs. We show that monocytes from IPF patients displayed increased expression of CD64 (FcγR1) which correlated with amount of lung fibrosis, and an amplified type I IFN response ex vivo. These were accompanied by markedly raised CSF-1 levels, IL-6, and CCL-2 in serum of IPF patients. Interrogation of single cell transcriptomic data from human IPF lungs revealed increased proportion of CD64hi monocytes and "transitional macrophages" with higher expression of CCL-2 and type I IFN genes. Our study shows that monocytes in IPF patients are phenotypically distinct from age-matched controls, with a primed type I IFN pathway that may contribute to driving chronic inflammation and fibrosis. These findings strengthen the potential role of monocytes in the pathogenesis of IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis/immunology , Interferon Type I/metabolism , Lung/immunology , Monocytes/immunology , Case-Control Studies , Cells, Cultured , Chemokine CCL2/blood , Flow Cytometry , Gene Expression Profiling , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Immunophenotyping , Interferon Type I/genetics , Interleukin-6/blood , Lung/metabolism , Lung/pathology , Macrophage Colony-Stimulating Factor/blood , Macrophages/immunology , Macrophages/metabolism , Monocytes/metabolism , Phenotype , Receptors, IgG/genetics , Receptors, IgG/metabolism , Single-Cell Analysis
17.
Hum Mol Genet ; 17(16): 2496-506, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18487244

ABSTRACT

We investigated clinical and cellular phenotypes of 24 children with mutations in the catalytic (alpha) subunit of the mitochondrial DNA (mtDNA) gamma polymerase (POLG1). Twenty-one had Alpers syndrome, the commonest severe POLG1 autosomal recessive phenotype, comprising hepatoencephalopathy and often mtDNA depletion. The cellular mtDNA content reflected the genotype more closely than did clinical features. Patients with tissue depletion of mtDNA all had at least one allele with either a missense mutation in a catalytic domain or a nonsense mutation. Four out of 12 patients exhibited a progressive, mosaic pattern of mtDNA depletion in cultured fibroblasts. All these patients had mutations in a catalytic domain in both POLG1 alleles, in either the polymerase or exonuclease domain or both. The tissue mtDNA content of patients who had two linker mutations was normal, and their phenotypes the mildest. Epilepsy and/or movement disorder were major features in all 21. Previous studies have implicated replication stalling as a mechanism for mtDNA depletion. The mosaic cellular depletion that we have demonstrated in cell cultures may be a manifestation of severe replication stalling. One patient with a severe cellular and clinical phenotype was a compound heterozygote with POLG1 mutations in the polymerase and exonuclease domain intrans. This suggests that POLG1 requires both polymerase and 3'-5' exonuclease activity in the same molecule. This is consistent with current functional models for eukaryotic DNA polymerases, which alternate between polymerizing and editing modes, as determined by competition between these two active sites for the 3' end of the DNA.


Subject(s)
DNA, Mitochondrial/genetics , DNA-Directed DNA Polymerase/genetics , Diffuse Cerebral Sclerosis of Schilder/enzymology , Diffuse Cerebral Sclerosis of Schilder/genetics , Fibroblasts/enzymology , Mutation , Adolescent , Cells, Cultured , Child , Child, Preschool , DNA Polymerase gamma , DNA, Mitochondrial/metabolism , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , Female , Fibroblasts/chemistry , Fibroblasts/cytology , Genotype , Humans , Infant , Life Expectancy , Male , Mitochondria/chemistry , Mitochondria/enzymology , Mitochondria/genetics , Phenotype , Protein Structure, Tertiary , Retrospective Studies
18.
Biochem Biophys Res Commun ; 378(3): 450-5, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19032935

ABSTRACT

The anthracyclines, such as doxorubicin (DXR), are potent anti-cancer drugs but they are limited by their clinical toxicity. The mechanisms involved remain poorly understood partly because of the difficulty in determining sub-cellular drug localisation. Using a novel method utilising the fluorescent DNA dye PicoGreen, we found that anthracyclines intercalated not only into nuclear DNA but also mitochondrial DNA (mtDNA). Intercalation of mtDNA by anthracyclines may thus contribute to the marked mitochondrial toxicity associated with these drugs. By contrast, ethidium bromide intercalated exclusively into mtDNA, without interacting with nuclear DNA, thereby explaining why mtDNA is the main target for ethidium. By exploiting PicoGreen quenching we also developed a novel assay for quantification of mtDNA levels by flow-cytometry, an approach which should be useful for studies of mitochondrial dysfunction. In summary our PicoGreen assay should be useful to study drug/DNA interactions within live cells, and facilitate therapeutic drug monitoring and kinetic studies in cancer patients.


Subject(s)
Anthracyclines/pharmacology , Antineoplastic Agents/pharmacology , DNA, Mitochondrial/drug effects , Intercalating Agents/pharmacology , Cell Line , Doxorubicin/pharmacology , Fluorescent Dyes/analysis , Humans , Organic Chemicals/analysis
19.
Sci Rep ; 9(1): 5300, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30923342

ABSTRACT

Priming haematopoietic stem/progenitor cells (HSPCs) in vitro with specific chromatin modifying agents and cytokines under serum-free-conditions significantly enhances engraftable HSC numbers. We extend these studies by culturing human CD133+ HSPCs on nanofibre scaffolds to mimic the niche for 5-days with the HDAC inhibitor Scriptaid and cytokines. Scriptaid increases absolute Lin-CD34+CD38-CD45RA-CD90+CD49f+ HSPC numbers, while concomitantly decreasing the Lin-CD38-CD34+CD45RA-CD90- subset. Hypothesising that Scriptaid plus cytokines expands the CD90+ subset without differentiation and upregulates CD90 on CD90- cells, we sorted, then cultured Lin-CD34+CD38-CD45RA-CD90- cells with Scriptaid and cytokines. Within 2-days and for at least 5-days, most CD90- cells became CD90+. There was no significant difference in the transcriptomic profile, by RNAsequencing, between cytokine-expanded and purified Lin-CD34+CD38-CD45RA-CD49f+CD90+ cells in the presence or absence of Scriptaid, suggesting that Scriptaid maintains stem cell gene expression programs despite expansion in HSC numbers. Supporting this, 50 genes were significantly differentially expressed between CD90+ and CD90- Lin-CD34+CD38-CD45RA-CD49f+ subsets in Scriptaid-cytokine- and cytokine only-expansion conditions. Thus, Scriptaid treatment of CD133+ cells may be a useful approach to expanding the absolute number of CD90+ HSC, without losing their stem cell characteristics, both through direct effects on HSC and potentially also conversion of their immediate CD90- progeny into CD90+ HSC.


Subject(s)
Hematologic Diseases/therapy , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/drug effects , Hydroxylamines/pharmacology , Quinolines/pharmacology , Transcriptome/drug effects , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Culture Media, Serum-Free , Fetal Blood/cytology , Hematopoietic Stem Cells/metabolism , Humans , RNA-Seq , Single-Cell Analysis , Thy-1 Antigens/genetics , Thy-1 Antigens/metabolism
20.
Nat Commun ; 10(1): 4533, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31586071

ABSTRACT

Multiple myeloma is an incurable, bone marrow-dwelling malignancy that disrupts bone homeostasis causing skeletal damage and pain. Mechanisms underlying myeloma-induced bone destruction are poorly understood and current therapies do not restore lost bone mass. Using transcriptomic profiling of isolated bone lining cell subtypes from a murine myeloma model, we find that bone morphogenetic protein (BMP) signalling is upregulated in stromal progenitor cells. BMP signalling has not previously been reported to be dysregulated in myeloma bone disease. Inhibition of BMP signalling in vivo using either a small molecule BMP receptor antagonist or a solubilized BMPR1a-FC receptor ligand trap prevents trabecular and cortical bone volume loss caused by myeloma, without increasing tumour burden. BMP inhibition directly reduces osteoclastogenesis, increases osteoblasts and bone formation, and suppresses bone marrow sclerostin levels. In summary we describe a novel role for the BMP pathway in myeloma-induced bone disease that can be therapeutically targeted.


Subject(s)
Bone Diseases/drug therapy , Bone Morphogenetic Proteins/metabolism , Multiple Myeloma/complications , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Stem Cells/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bone Density/drug effects , Bone Diseases/etiology , Bone Diseases/pathology , Bone Marrow/pathology , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Protein Receptors/metabolism , Cell Line, Tumor , Disease Models, Animal , Femur/cytology , Femur/drug effects , Femur/pathology , Gene Expression Profiling , Gene Expression Regulation , Humans , Injections, Intraperitoneal , Mice , Mice, Inbred Strains , Multiple Myeloma/pathology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , RNA-Seq , Signal Transduction/drug effects , Stem Cells/pathology , Tibia/cytology , Tibia/drug effects , Tibia/pathology , Treatment Outcome , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL