Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Nat Immunol ; 21(12): 1563-1573, 2020 12.
Article in English | MEDLINE | ID: mdl-33106669

ABSTRACT

Chronic cytomegalovirus (CMV) infection leads to long-term maintenance of extraordinarily large CMV-specific T cell populations. The magnitude of this so-called 'memory inflation' is thought to mainly depend on antigenic stimulation during the chronic phase of infection. However, by mapping the long-term development of CD8+ T cell families derived from single naive precursors, we find that fate decisions made during the acute phase of murine CMV infection can alter the level of memory inflation by more than 1,000-fold. Counterintuitively, a T cell family's capacity for memory inflation is not determined by its initial expansion. Instead, those rare T cell families that dominate the chronic phase of infection show an early transcriptomic signature akin to that of established T central memory cells. Accordingly, a T cell family's long-term dominance is best predicted by its early content of T central memory precursors, which later serve as a stem-cell-like source for memory inflation.


Subject(s)
Clonal Evolution/immunology , Host-Pathogen Interactions/immunology , Immunologic Memory , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Virus Diseases/etiology , Virus Diseases/metabolism , Acute Disease , Animals , Biomarkers , Chronic Disease , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Gene Expression Profiling , Humans , Immunophenotyping , Mice , Muromegalovirus/immunology
2.
Immunity ; 54(10): 2288-2304.e7, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34437840

ABSTRACT

Upon viral infection, natural killer (NK) cells expressing certain germline-encoded receptors are selected, expanded, and maintained in an adaptive-like manner. Currently, these are thought to differentiate along a common pathway. However, by fate mapping of single NK cells upon murine cytomegalovirus (MCMV) infection, we identified two distinct NK cell lineages that contributed to adaptive-like responses. One was equivalent to conventional NK (cNK) cells while the other was transcriptionally similar to type 1 innate lymphoid cells (ILC1s). ILC1-like NK cells showed splenic residency and strong cytokine production but also recognized and killed MCMV-infected cells, guided by activating receptor Ly49H. Moreover, they induced clustering of conventional type 1 dendritic cells and facilitated antigen-specific T cell priming early during MCMV infection, which depended on Ly49H and the NK cell-intrinsic expression of transcription factor Batf3. Thereby, ILC1-like NK cells bridge innate and adaptive viral recognition and unite critical features of cNK cells and ILC1s.


Subject(s)
Adaptive Immunity/immunology , Cell Lineage/immunology , Herpesviridae Infections/immunology , Immunity, Innate/immunology , Killer Cells, Natural/immunology , Animals , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muromegalovirus
3.
Immunity ; 50(6): 1391-1400.e4, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31103380

ABSTRACT

Natural killer (NK) cells show some features of adaptive immunity but have not been studied at the clonal level. Here, we used retrogenic color-barcoding and single-cell adoptive transfers to track clonal immune responses to murine cytomegalovirus (MCMV) infection, derived from individual NK cells expressing activating receptor Ly49H. Clonal expansion of single NK cells varied substantially, and this variation could not be attributed to the additional presence or absence of inhibitory Ly49 receptors. Instead, single-cell-derived variability correlated with distinct surface expression levels of Ly49H itself. Ly49Hhi NK cell clones maintained higher Ly49H expression and expanded more than their Ly49Hlo counterparts in response to MCMV. Thus, akin to adaptive processes shaping an antigen-specific T cell receptor (TCR) repertoire, the Ly49H+ NK cell population adapts to MCMV infection. This process relies on the clonal maintenance of distinct Ly49H expression levels, generating a repertoire of individual NK cells outfitted with distinct reactivity to MCMV.


Subject(s)
Cytomegalovirus Infections , Muromegalovirus , Animals , Killer Cells, Natural , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily A
4.
Blood ; 139(5): 690-703, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34657154

ABSTRACT

The cellular mechanisms required to ensure homeostasis of the hematopoietic niche and the ability of this niche to support hematopoiesis upon stress remain elusive. We here identify Wnt5a in Osterix+ mesenchymal progenitor and stem cells (MSPCs) as a critical factor for niche-dependent hematopoiesis. Mice lacking Wnt5a in MSPCs suffer from stress-related bone marrow (BM) failure and increased mortality. Niche cells devoid of Wnt5a show defective actin stress fiber orientation due to an elevated activity of the small GTPase CDC42. This results in incorrect positioning of autophagosomes and lysosomes, thus reducing autophagy and increasing oxidative stress. In MSPCs from patients from BM failure states which share features of peripheral cytopenia and hypocellular BM, we find similar defects in actin stress fiber orientation, reduced and incorrect colocalization of autophagosomes and lysosomes, and CDC42 activation. Strikingly, a short pharmacological intervention to attenuate elevated CDC42 activation in vivo in mice prevents defective actin-anchored autophagy in MSPCs, salvages hematopoiesis and protects against lethal cytopenia upon stress. In summary, our study identifies Wnt5a as a restriction factor for niche homeostasis by affecting CDC42-regulated actin stress-fiber orientation and autophagy upon stress. Our data further imply a critical role for autophagy in MSPCs for adequate support of hematopoiesis by the niche upon stress and in human diseases characterized by peripheral cytopenias and hypocellular BM.


Subject(s)
Autophagy , Bone Marrow Failure Disorders/metabolism , Hematopoiesis , Mesenchymal Stem Cells/cytology , Animals , Cells, Cultured , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Mice , Oxidative Stress , Wnt-5a Protein/metabolism
5.
Immunity ; 41(1): 116-26, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25035956

ABSTRACT

Maintenance of immunological memory has been proposed to rely on stem-cell-like lymphocytes. However, data supporting this hypothesis are focused on the developmental potential of lymphocyte populations and are thus insufficient to establish the functional hallmarks of stemness. Here, we investigated self-renewal capacity and multipotency of individual memory lymphocytes by in vivo fate mapping of CD8(+) T cells and their descendants across three generations of serial single-cell adoptive transfer and infection-driven re-expansion. We found that immune responses derived from single naive T (Tn) cells, single primary, and single secondary central memory T (Tcm) cells reached similar size and phenotypic diversity, were subjected to comparable stochastic variation, and could ultimately reconstitute immunocompetence against an otherwise lethal infection with the bacterial pathogen Listeria monocytogenes. These observations establish that adult tissue stem cells reside within the CD62L(+) Tcm cell compartment and highlight the promising therapeutic potential of this immune cell subset.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Lineage/immunology , Immunologic Memory/immunology , Adult Stem Cells/immunology , Animals , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/immunology , Immunocompetence/immunology , Immunotherapy, Adoptive , L-Selectin/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multipotent Stem Cells/immunology , T-Lymphocyte Subsets/immunology
6.
Haematologica ; 106(10): 2633-2640, 2021 10 01.
Article in English | MEDLINE | ID: mdl-33543864

ABSTRACT

Hematopoietic stem cell self-renewal, proliferation, and differentiation are independently regulated by intrinsic as well as extrinsic mechanisms. We previously demonstrated that murine proliferation of hematopoietic stem cells is supported in serum-free medium supplemented with two growth factors, stem cell factor and interleukin 11. The survival of hematopoietic stem cells is additionally improved by supplementing this medium with two more growth factors, neural growth factor and collagen 1 (four growth factors) or serum-free medium conditioned by the hematopoietic stem cell-supportive stromal UG26-1B6 cells1. Here, we describe a robust and versatile alternative source of conditioned medium from mouse embryonic fibroblasts. We found that this conditioned medium supports survival and phenotypical identity of hematopoietic stem cells, as well as cell cycle entry in single cell cultures of CD34- CD48- CD150+ Lineage- SCA1+ KIT+ cells supplemented with two growth factors. Strikingly, in comparison with cultures in serum-free medium with four growth factors, conditioned medium from mouse embryonic fibroblasts increases the numbers of proliferating clones and the number of Lineage- SCA1+ KIT+ cells, both with two and four growth factors. In addition, conditioned medium from mouse embryonic fibroblasts supports self-renewal in culture of cells with short- and long-term hematopoiesis-repopulating ability in vivo. These findings identify conditioned medium from mouse embryonic fibroblasts as a robust alternative serumfree source of factors to maintain self-renewal of in vivo-repopulating hematopoetic stem cells in culture.


Subject(s)
Fibroblasts , Hematopoietic Stem Cells , Animals , Cell Differentiation , Cell Division , Cells, Cultured , Hematopoiesis , Mice
7.
Cytometry A ; 97(2): 171-183, 2020 02.
Article in English | MEDLINE | ID: mdl-31944553

ABSTRACT

Cell alterations during isolation and preparation for flow cytometry cell sorting by antibodies, temperature, homogenization, buffer composition and mitogens are well known. In contrast, little is known about cell alteration caused by the instrument or the sorting process itself. We systematically evaluated cellular responses to different sorter-induced physical forces. In summary, flow cytometry cell-sorting induced forces can affect cellular signaling cascades, especially the MAPK p38. Functional assays, related to the p38 MAPK pathway, of human primary T cells after flow cytometry sorting did lead to minor physiological modulation but no functional impairments. © 2020 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.


Subject(s)
T-Lymphocytes , p38 Mitogen-Activated Protein Kinases , Cell Separation , Flow Cytometry , Humans , Signal Transduction , T-Lymphocytes/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Nature ; 503(7476): 392-6, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24141946

ABSTRACT

Many organs with a high cell turnover (for example, skin, intestine and blood) are composed of short-lived cells that require continuous replenishment by somatic stem cells. Ageing results in the inability of these tissues to maintain homeostasis and it is believed that somatic stem-cell ageing is one underlying cause of tissue attrition with age or age-related diseases. Ageing of haematopoietic stem cells (HSCs) is associated with impaired haematopoiesis in the elderly. Despite a large amount of data describing the decline of HSC function on ageing, the molecular mechanisms of this process remain largely unknown, which precludes rational approaches to attenuate stem-cell ageing. Here we report an unexpected shift from canonical to non-canonical Wnt signalling in mice due to elevated expression of Wnt5a in aged HSCs, which causes stem-cell ageing. Wnt5a treatment of young HSCs induces ageing-associated stem-cell apolarity, reduction of regenerative capacity and an ageing-like myeloid-lymphoid differentiation skewing via activation of the small Rho GTPase Cdc42. Conversely, Wnt5a haploinsufficiency attenuates HSC ageing, whereas stem-cell-intrinsic reduction of Wnt5a expression results in functionally rejuvenated aged HSCs. Our data demonstrate a critical role for stem-cell-intrinsic non-canonical Wnt5a signalling in HSC ageing.


Subject(s)
Cellular Senescence , Hematopoietic Stem Cells/cytology , Wnt Signaling Pathway , Animals , Cell Differentiation , Cell Polarity , Female , Haploinsufficiency , Male , Mice , Mice, Inbred C57BL , Phenotype , Rejuvenation , Wnt Proteins/deficiency , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt-5a Protein , cdc42 GTP-Binding Protein/metabolism
9.
J Virol ; 90(20): 8954-67, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27384654

ABSTRACT

UNLABELLED: Interleukin 2 (IL-2) signaling through the IL-2 receptor alpha chain (CD25) facilitates HIV replication in vitro and facilitates homeostatic proliferation of CD25(+) FoxP3(+) CD4(+) T cells. CD25(+) FoxP3(+) CD4(+) T cells may therefore constitute a suitable subset for HIV infection and plasma virion production. CD25(+) FoxP3(+) CD4(+) T cell frequencies, absolute numbers, and the expression of CCR5 and cell cycle marker Ki67 were studied in peripheral blood from HIV(+) and HIV(-) study volunteers. Different memory CD4(+) T cell subsets were then sorted for quantification of cell-associated HIV DNA and phylogenetic analyses of the highly variable EnvV1V3 region in comparison to plasma-derived virus sequences. In HIV(+) subjects, 51% (median) of CD25(+) FoxP3(+) CD4(+) T cells expressed the HIV coreceptor CCR5. Very high frequencies of Ki67(+) cells were detected in CD25(+) FoxP3(+) memory CD4(+) T cells (median, 27.6%) in comparison to CD25(-) FoxP3(-) memory CD4(+) T cells (median, 4.1%; P < 0.0001). HIV DNA content was 15-fold higher in CD25(+) FoxP3(+) memory CD4(+) T cells than in CD25(-) FoxP3(-) T cells (P = 0.003). EnvV1V3 sequences derived from CD25(+) FoxP3(+) memory CD4(+) T cells did not preferentially cluster with plasma-derived sequences. Quasi-identical cell-plasma sequence pairs were rare, and their proportion decreased with the estimated HIV infection duration. These data suggest that specific cellular characteristics of CD25(+) FoxP3(+) memory CD4(+) T cells might facilitate efficient HIV infection in vivo and passage of HIV DNA to cell progeny in the absence of active viral replication. The contribution of this cell population to plasma virion production remains unclear. IMPORTANCE: Despite recent advances in the understanding of AIDS virus pathogenesis, which cell subsets support HIV infection and replication in vivo is incompletely understood. In vitro, the IL-2 signaling pathway and IL-2-dependent cell cycle induction are essential for HIV infection of stimulated T cells. CD25(+) FoxP3(+) memory CD4 T cells, often referred to as regulatory CD4 T cells, depend on IL-2 signaling for homeostatic proliferation in vivo Our results show that CD25(+) FoxP3(+) memory CD4(+) T cells often express the HIV coreceptor CCR5, are significantly more proliferative, and contain more HIV DNA than CD25(-) FoxP3(-) memory CD4 T cell subsets. The specific cellular characteristics of CD25(+) FoxP3(+) memory CD4(+) T cells probably facilitate efficient HIV infection in vivo and passage of HIV DNA to cell progeny in the absence of active viral replication. However, the contribution of this cell subset to plasma viremia remains unclear.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Forkhead Transcription Factors/analysis , HIV Infections/virology , HIV/isolation & purification , Interleukin-2 Receptor alpha Subunit/analysis , Receptors, CCR5/analysis , T-Lymphocyte Subsets/virology , CD4-Positive T-Lymphocytes/chemistry , DNA, Viral/analysis , DNA, Viral/genetics , HIV/classification , HIV/genetics , Humans , Ki-67 Antigen/analysis , Phylogeny , Sequence Analysis, DNA , T-Lymphocyte Subsets/chemistry , env Gene Products, Human Immunodeficiency Virus/genetics
10.
J Immunol ; 195(4): 1753-62, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26170386

ABSTRACT

Plasmacytoid dendritic cells (pDCs) efficiently produce large amounts of type I IFN in response to TLR7 and TLR9 ligands, whereas conventional DCs (cDCs) predominantly secrete high levels of the cytokines IL-10 and IL-12. The molecular basis underlying this distinct phenotype is not well understood. In this study, we identified the MAPK phosphatase Dusp9/MKP-4 by transcriptome analysis as selectively expressed in pDCs, but not cDCs. We confirmed the constitutive expression of Dusp9 at the protein level in pDCs generated in vitro by culture with Flt3 ligand and ex vivo in sorted splenic pDCs. Dusp9 expression was low in B220(-) bone marrow precursors and was upregulated during pDC differentiation, concomitant with established pDC markers. Higher expression of Dusp9 in pDCs correlated with impaired phosphorylation of the MAPK ERK1/2 upon TLR9 stimulation. Notably, Dusp9 was not expressed at detectable levels in human pDCs, although these displayed similarly impaired activation of ERK1/2 MAPK compared with cDCs. Enforced retroviral expression of Dusp9 in mouse GM-CSF-induced cDCs increased the expression of TLR9-induced IL-12p40 and IFN-ß, but not of IL-10. Conditional deletion of Dusp9 in pDCs was effectively achieved in Dusp9(flox/flox); CD11c-Cre mice at the mRNA and protein levels. However, the lack of Dusp9 in pDC did not restore ERK1/2 activation after TLR9 stimulation and only weakly affected IFN-ß and IL-12p40 production. Taken together, our results suggest that expression of Dusp9 is sufficient to impair ERK1/2 activation and enhance IFN-ß expression. However, despite selective expression in pDCs, Dusp9 is not essential for high-level IFN-ß production by these cells.


Subject(s)
Dendritic Cells/metabolism , Dual-Specificity Phosphatases/genetics , Gene Expression , Interferon-beta/biosynthesis , Animals , Cell Differentiation/genetics , Cluster Analysis , Computational Biology/methods , Dendritic Cells/cytology , Dendritic Cells/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Profiling , Humans , Interferon-beta/genetics , Interleukin-12 , Mice , Mice, Knockout , Organ Specificity/genetics , Phosphorylation , Reproducibility of Results , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/metabolism , Transcriptome
11.
Gastroenterology ; 149(4): 1042-52, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26052074

ABSTRACT

BACKGROUND & AIMS: Cancer therapies are being developed based on our ability to direct T cells against tumor antigens. Glypican-3 (GPC3) is expressed by 75% of all hepatocellular carcinomas (HCC), but not in healthy liver tissue or other organs. We aimed to generate T cells with GPC3-specific receptors that recognize HCC and used them to eliminate GPC3-expressing xenograft tumors grown from human HCC cells in mice. METHODS: We used mass spectrometry to obtain a comprehensive peptidome from GPC3-expressing hepatoma cells after immune-affinity purification of human leukocyte antigen (HLA)-A2 and bioinformatics to identify immunodominant peptides. To circumvent GPC3 tolerance resulting from fetal expression, dendritic cells from HLA-A2-negative donors were cotransfected with GPC3 and HLA-A2 RNA to stimulate and expand antigen-specific T cells. RESULTS: Peptide GPC3367 was identified as a predominant peptide on HLA-A2. We used A2-GPC3367 multimers to detect, select for, and clone GPC3-specific T cells. These clones bound the A2-GPC3367 multimer and secreted interferon-γ when cultured with GPC3367, but not with control peptide-loaded cells. By genomic sequencing of these T-cell clones, we identified a gene encoding a dominant T-cell receptor. The gene was cloned and the sequence was codon optimized and expressed from a retroviral vector. Primary CD8(+) T cells that expressed the transgenic T-cell receptor specifically bound GPC3367 on HLA-A2. These T cells killed GPC3-expressing hepatoma cells in culture and slowed growth of HCC xenograft tumors in mice. CONCLUSIONS: We identified a GPC3367-specific T-cell receptor. Expression of this receptor by T cells allows them to recognize and kill GPC3-positive hepatoma cells. This finding could be used to advance development of adoptive T-cell therapy for HCC.


Subject(s)
CD8-Positive T-Lymphocytes/transplantation , Carcinoma, Hepatocellular/therapy , Cytotoxicity, Immunologic , Dendritic Cells/metabolism , Genes, T-Cell Receptor , Genetic Engineering/methods , Glypicans/metabolism , HLA-A2 Antigen/metabolism , Immunotherapy, Adoptive/methods , Liver Neoplasms/therapy , Lymphocyte Activation , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Survival , Coculture Techniques , Dendritic Cells/immunology , Female , Glypicans/genetics , Glypicans/immunology , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Hep G2 Cells , Humans , Immunodominant Epitopes , Interferon-gamma/immunology , Interferon-gamma/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice, SCID , Time Factors , Transfection , Xenograft Model Antitumor Assays
12.
Cytometry A ; 89(9): 816-25, 2016 09.
Article in English | MEDLINE | ID: mdl-27564267

ABSTRACT

High epitope-specific sensitivity of CD8(+) T cells is required for optimal immune protection against intracellular pathogens as well as certain malignancies. The quality of antigen recognition of CD8(+) T cells is usually described as "avidity" to its cognate peptide MHCI complex. T cell avidity is mainly dependent on the structural qualities of the T cell receptor (TCR), as convincingly demonstrated by recombinant TCR re-expression experiments. Based on reversible MHCI multimer staining and koff -rate measurements of monomeric peptide MHCI complexes, we recently established a microscopic assay for determining the structural avidity of individual CD8(+) T cells. Here we demonstrate that this assay can be adapted for rapid flow-cytometric avidity screening of epitope-specific T cell populations. Furthermore, we show that-in combination with conventional nonreversible MHCI multimer staining-even very small epitope-specific CD8(+) T cell populations can be analyzed directly ex vivo without the need for previous TCR cloning or T cell sorting. This simplified approach provides highly accurate mean TCR-ligand koff -rate values for poly- or oligoclonal T cell populations and is ideally suited for high-throughput applications in basic research as well as clinical settings. © 2016 International Society for Advancement of Cytometry.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Flow Cytometry/methods , T-Lymphocytes/immunology , Animals , Antigens/immunology , Humans , Ligands , Lymphocyte Activation/immunology , Mice , Receptors, Antigen, T-Cell/immunology
13.
Blood ; 124(4): 628-37, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-24855206

ABSTRACT

Patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) are threatened by potentially lethal viral manifestations like cytomegalovirus (CMV) reactivation. Because the success of today's virostatic treatment is limited by side effects and resistance development, adoptive transfer of virus-specific memory T cells derived from the stem cell donor has been proposed as an alternative therapeutic strategy. In this context, dose minimization of adoptively transferred T cells might be warranted for the avoidance of graft-versus-host disease (GVHD), in particular in prophylactic settings after T-cell-depleting allo-HSCT protocols. To establish a lower limit for successful adoptive T-cell therapy, we conducted low-dose CD8(+) T-cell transfers in the well-established murine Listeria monocytogenes (L.m.) infection model. Major histocompatibility complex-Streptamer-enriched antigen-specific CD62L(hi) but not CD62L(lo) CD8(+) memory T cells proliferated, differentiated, and protected against L.m. infections after prophylactic application. Even progenies derived from a single CD62L(hi) L.m.-specific CD8(+) T cell could be protective against bacterial challenge. In analogy, low-dose transfers of Streptamer-enriched human CMV-specific CD8(+) T cells into allo-HSCT recipients led to strong pathogen-specific T-cell expansion in a compassionate-use setting. In summary, low-dose adoptive T-cell transfer (ACT) could be a promising strategy, particularly for prophylactic treatment of infectious complications after allo-HSCT.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Graft vs Host Disease/immunology , Immunotherapy, Adoptive , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Severe Combined Immunodeficiency/immunology , Adolescent , Animals , Cell Differentiation , Cell Proliferation , Child , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/therapy , Graft vs Host Disease/metabolism , Graft vs Host Disease/therapy , Hematopoietic Stem Cell Transplantation , Homeodomain Proteins/physiology , Humans , Immunization , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/physiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Severe Combined Immunodeficiency/metabolism , Severe Combined Immunodeficiency/therapy , Transplantation, Homologous , Virus Activation
14.
Stem Cells ; 33(4): 1113-29, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25524439

ABSTRACT

During cardiogenesis, most myocytes arise from cardiac progenitors expressing the transcription factors Isl1 and Nkx2-5. Here, we show that a direct repression of Isl1 by Nkx2-5 is necessary for proper development of the ventricular myocardial lineage. Overexpression of Nkx2-5 in mouse embryonic stem cells (ESCs) delayed specification of cardiac progenitors and inhibited expression of Isl1 and its downstream targets in Isl1(+) precursors. Embryos deficient for Nkx2-5 in the Isl1(+) lineage failed to downregulate Isl1 protein in cardiomyocytes of the heart tube. We demonstrated that Nkx2-5 directly binds to an Isl1 enhancer and represses Isl1 transcriptional activity. Furthermore, we showed that overexpression of Isl1 does not prevent cardiac differentiation of ESCs and in Xenopus laevis embryos. Instead, it leads to enhanced specification of cardiac progenitors, earlier cardiac differentiation, and increased cardiomyocyte number. Functional and molecular characterization of Isl1-overexpressing cardiomyocytes revealed higher beating frequencies in both ESC-derived contracting areas and Xenopus Isl1-gain-of-function hearts, which associated with upregulation of nodal-specific genes and downregulation of transcripts of working myocardium. Immunocytochemistry of cardiomyocyte lineage-specific markers demonstrated a reduction of ventricular cells and an increase of cells expressing the pacemaker channel Hcn4. Finally, optical action potential imaging of single cardiomyocytes combined with pharmacological approaches proved that Isl1 overexpression in ESCs resulted in normally electrophysiologically functional cells, highly enriched in the nodal subtype at the expense of the ventricular lineage. Our findings provide an Isl1/Nkx2-5-mediated mechanism that coordinately regulates the specification of cardiac progenitors toward the different myocardial lineages and ensures proper acquisition of myocyte subtype identity.


Subject(s)
Homeodomain Proteins/biosynthesis , LIM-Homeodomain Proteins/antagonists & inhibitors , LIM-Homeodomain Proteins/biosynthesis , Myocytes, Cardiac/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis , Animals , Cell Lineage/physiology , Embryonic Stem Cells/metabolism , HEK293 Cells , Homeobox Protein Nkx-2.5 , Humans , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Protein Binding/physiology , Xenopus
15.
Stem Cells ; 33(2): 392-402, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25335772

ABSTRACT

The generation of induced pluripotent stem (iPS) cells has successfully been achieved in many species. However, the identification of truly reprogrammed iPS cells still remains laborious and the detection of pluripotency markers requires fixation of cells in most cases. Here, we report an approach with nanoparticles carrying Cy3-labeled sense oligonucleotide reporter strands coupled to gold-particles. These molecules are directly added to cultured cells without any manipulation and gene expression is evaluated microscopically after overnight incubation. To simultaneously detect gene expression in different species, probe sequences were chosen according to interspecies homology. With a common target-specific probe we could successfully demonstrate expression of the GAPDH house-keeping gene in somatic cells and expression of the pluripotency markers NANOG and GDF3 in embryonic stem cells and iPS cells of murine, human, and porcine origin. The population of target gene positive cells could be purified by fluorescence-activated cell sorting. After lentiviral transduction of murine tail-tip fibroblasts Nanog-specific probes identified truly reprogrammed murine iPS cells in situ during development based on their Cy3-fluorescence. The intensity of Nanog-specific fluorescence correlated positively with an increased capacity of individual clones to differentiate into cells of all three germ layers. Our approach offers a universal tool to detect intracellular gene expression directly in live cells of any desired origin without the need for manipulation, thus allowing conservation of the genetic background of the target cell. Furthermore, it represents an easy, scalable method for efficient screening of pluripotency which is highly desirable during high-throughput cell reprogramming and after genomic editing of pluripotent stem cells.


Subject(s)
Carbocyanines/pharmacology , Cellular Reprogramming , Embryonic Stem Cells/metabolism , Gene Expression Regulation , Induced Pluripotent Stem Cells/metabolism , Oligonucleotides/pharmacology , Animals , Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Mice, Transgenic , Swine
16.
Genomics ; 106(1): 7-14, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25812950

ABSTRACT

UNLABELLED: Pancreatic cancer is one of the most aggressive malignant tumors, mainly due to an aggressive metastasis spreading. In recent years, circulating tumor cells became associated to tumor metastasis. Little is known about their expression profiles. The aim of this study was to develop a complete workflow making it possible to isolate circulating tumor cells from patients with pancreatic cancer and their genetic characterization. RESULTS: We show that the proposed workflow offers a technical sensitivity and specificity high enough to detect and isolate single tumor cells. Moreover our approach makes feasible to genetically characterize single CTCs. CONCLUSIONS: Our work discloses a complete workflow to detect, count and genetically analyze individual CTCs isolated from blood samples. This method has a central impact on the early detection of metastasis development. The combination of cell quantification and genetic analysis provides the clinicians with a powerful tool not available so far.


Subject(s)
Neoplastic Cells, Circulating/metabolism , Pancreatic Neoplasms/genetics , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Separation , Female , Flow Cytometry , Humans , Male , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pilot Projects
17.
Hepatology ; 57(6): 2358-68, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23424039

ABSTRACT

UNLABELLED: Alternatively polarized macrophages (Mϕ) shape the microenvironment of hepatocellular carcinoma (HCC) and temper anticancer immune responses. We investigated if sorafenib alters the HCC microenvironment by restoring classical macrophage polarization and triggering tumor-directed natural killer (NK) cell responses. In vivo experiments were conducted with sorafenib (25 mg/kg)-treated C57BL/6 wildtype as well as hepatitis B virus (HBV) and lymphotoxin transgenic mice with and without HCC. Monocyte-derived Mϕ or tumor-associated macrophages (TAM) isolated from HCC tissue were treated with sorafenib (0.07-5.0 µg/mL) and cocultured with autologous NK cells. Mϕ and NK cell activation was analyzed by flow cytometry and killing assays, respectively. Cytokine and growth factor release was measured by enzyme-linked immunosorbent assay. Short-term administration of sorafenib triggered activation of hepatic NK cells in wildtype and tumor-bearing mice. In vitro, sorafenib sensitized Mϕ to lipopolysaccharide, reverted alternative Mϕ polarization and enhanced IL12 secretion (P = 0.0133). NK cells activated by sorafenib-treated Mϕ showed increased degranulation (15.3 ± 0.2% versus 32.0 ± 0.9%, P < 0.0001) and interferon-gamma (IFN-γ) secretion (2.1 ± 0.2% versus 8.0 ± 0.2%, P < 0.0001) upon target cell contact. Sorafenib-triggered NK cell activation was verified by coculture experiments using TAM. Sorafenib-treated Mϕ increased cytolytic NK cell function against K562, Raji, and HepG2 target cells in a dose-dependent manner. Neutralization of interleukin (IL)12 or IL18 as well as inhibition of the nuclear factor kappa B (NF-κB) pathway reversed NK cell activation in Mϕ/NK cocultures. CONCLUSION: Sorafenib triggers proinflammatory activity of TAM and subsequently induces antitumor NK cell responses in a cytokine- and NF-κB-dependent fashion. This observation is relevant for HCC therapy, as sorafenib is a compound in clinical use that reverts alternative polarization of TAM in HCC. (HEPATOLOGY 2013;57:2358-2368).


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Macrophage Activation/drug effects , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Carcinoma, Hepatocellular/immunology , Cytokines/metabolism , Drug Evaluation, Preclinical , Humans , Killer Cells, Natural/drug effects , Liver Neoplasms/immunology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Niacinamide/pharmacology , Niacinamide/therapeutic use , Phenylurea Compounds/therapeutic use , Sorafenib
18.
Blood ; 119(25): 6063-71, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22547585

ABSTRACT

The ontogenic relationship between the common dendritic cell (DC) progenitor (CDP), the committed conventional DC precursor (pre-cDC), and cDC subpopulations in lymphoid and nonlymphoid tissues has been largely unraveled. In contrast, the sequential steps of plasmacytoid DC (pDC) development are less defined, and it is unknown at which developmental stage and location final commitment to the pDC lineage occurs. Here we show that CCR9(-) pDCs from murine BM which enter the circulation and peripheral tissues have a common DC precursor function in vivo in the steady state, in contrast to CCR9(+) pDCs which are terminally differentiated. On adoptive transfer, the fate of CCR9(-) pDC-like precursors is governed by the tissues they enter. In the BM and liver, most transferred CCR9(-) pDC-like precursors differentiate into CCR9(+) pDCs, whereas in peripheral lymphoid organs, lung, and intestine, they additionally give rise to cDCs. CCR9(-) pDC-like precursors which are distinct from pre-cDCs can be generated from the CDP. Thus, CCR9(-) pDC-like cells are novel CDP-derived circulating DC precursors with pDC and cDC potential. Their final differentiation into functionally distinct pDCs and cDCs depends on tissue-specific factors allowing adaptation to local requirements under homeostatic conditions.


Subject(s)
Cell Differentiation , Dendritic Cells/physiology , Stem Cells/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/genetics , Cells, Cultured , Dendritic Cells/metabolism , Female , Gene Expression Profiling , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Lymphoid Tissue/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucous Membrane/cytology , Mucous Membrane/metabolism , Mucous Membrane/physiology , Organ Specificity/genetics , Receptors, CCR/metabolism , Specific Pathogen-Free Organisms , Stem Cells/metabolism
19.
Stem Cells ; 31(8): 1467-76, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23630186

ABSTRACT

In many solid tumors, cancer stem cells (CSC) represent a population with tumor-initiating, self-renewal, and differentiation potential, which can be identified by surface protein markers. No generally applicable markers are yet known for renal cell carcinoma (RCC). Two RCC cell lines (RCC-26, RCC-53) were found to differ widely in their capacity to form spheres in vitro and to establish tumors in mice, potentially reflecting differences in CSC content. A subpopulation expressing the CXC chemokine receptor 4 (CXCR4) was present only in the more tumorigenic cell line RCC-53. When grown as spheres, most of the RCC-53 cells were CXCR4-positive, expressed stem cell-associated transcription factor genes at elevated levels, and were more resistant toward the tyrosine kinase inhibitors sunitinib, sorafenib, and pazopanib. Sorted CXCR4-positive cells exhibited greater capacity for sphere formation and tumor growth-inducing potential in vivo than CXCR4-negative cells. Significantly, higher CXCR4 mRNA levels in primary RCC tumors from patients with localized but not disseminated disease predicted shorter survival. Downregulation of CXCR4 expression by small interfering RNA (siRNA) or pharmacological inhibition by AMD3100 compromised tumor sphere formation, viability of CXCR4-positive cells, and increased their responsiveness toward tyrosine kinase inhibitors. In conclusion, CXCR4 identifies a subpopulation of tumor-initiating cells in RCC cell lines and plays a role in their maintenance. The relative insensitivity of such cells to tyrosine kinase inhibitors might contribute to the development of therapy resistance in RCC patients. Future therapies therefore could combine blockade of the CXCR4 signaling pathway with standard therapies for more effective treatments of metastatic RCC.


Subject(s)
Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Receptors, CXCR4/metabolism , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Movement/physiology , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Heterografts , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Neoplastic Stem Cells/pathology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Receptors, CXCR4/genetics , Signal Transduction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL