Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Cell ; 186(3): 621-645.e33, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36736301

ABSTRACT

Inborn errors of human IFN-γ-dependent macrophagic immunity underlie mycobacterial diseases, whereas inborn errors of IFN-α/ß-dependent intrinsic immunity underlie viral diseases. Both types of IFNs induce the transcription factor IRF1. We describe unrelated children with inherited complete IRF1 deficiency and early-onset, multiple, life-threatening diseases caused by weakly virulent mycobacteria and related intramacrophagic pathogens. These children have no history of severe viral disease, despite exposure to many viruses, including SARS-CoV-2, which is life-threatening in individuals with impaired IFN-α/ß immunity. In leukocytes or fibroblasts stimulated in vitro, IRF1-dependent responses to IFN-γ are, both quantitatively and qualitatively, much stronger than those to IFN-α/ß. Moreover, IRF1-deficient mononuclear phagocytes do not control mycobacteria and related pathogens normally when stimulated with IFN-γ. By contrast, IFN-α/ß-dependent intrinsic immunity to nine viruses, including SARS-CoV-2, is almost normal in IRF1-deficient fibroblasts. Human IRF1 is essential for IFN-γ-dependent macrophagic immunity to mycobacteria, but largely redundant for IFN-α/ß-dependent antiviral immunity.


Subject(s)
COVID-19 , Mycobacterium , Child , Humans , Interferon-gamma , SARS-CoV-2 , Interferon-alpha , Interferon Regulatory Factor-1
2.
Haematologica ; 106(5): 1354-1367, 2021 05 01.
Article in English | MEDLINE | ID: mdl-32327499

ABSTRACT

Hematopoietic development is spatiotemporally tightly regulated by defined cell-intrinsic and extrinsic modifiers. The role of cytokines has been intensively studied in adult hematopoiesis; however, their role in embryonic hematopoietic specification remains largely unexplored. Here, we used induced pluripotent stem cell (iPSC) technology and established a 3-dimensional, organoid-like differentiation system (hemanoid) maintaining the structural cellular integrity to evaluate the effect of cytokines on embryonic hematopoietic development. We show, that defined stages of early human hematopoietic development were recapitulated within the generated hemanoids. We identified KDR+/CD34high/CD144+/CD43-/CD45- hemato-endothelial progenitor cells (HEPs) forming organized, vasculature-like structures and giving rise to CD34low/CD144-/CD43+/CD45+ hematopoietic progenitor cells. We demonstrate that the endothelial to hematopoietic transition of HEPs is dependent on the presence of interleukin 3 (IL-3). Inhibition of IL-3 signalling blocked hematopoietic differentiation and arrested the cells in the HEP stage. Thus, our data suggest an important role for IL-3 in early human hematopoiesis by supporting the endothelial to hematopoietic transition of hemato-endothelial progenitor cells and highlight the potential of a hemanoid-based model to study human hematopoietic development.


Subject(s)
Induced Pluripotent Stem Cells , Interleukin-3 , Pluripotent Stem Cells , Adult , Cell Differentiation , Hematopoiesis , Humans
3.
J Exp Med ; 218(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34264265

ABSTRACT

Patients with autosomal recessive protein kinase C δ (PKCδ) deficiency suffer from childhood-onset autoimmunity, including systemic lupus erythematosus. They also suffer from recurrent infections that overlap with those seen in patients with chronic granulomatous disease (CGD), a disease caused by defects of the phagocyte NADPH oxidase and a lack of reactive oxygen species (ROS) production. We studied an international cohort of 17 PKCδ-deficient patients and found that their EBV-B cells and monocyte-derived phagocytes produced only small amounts of ROS and did not phosphorylate p40phox normally after PMA or opsonized Staphylococcus aureus stimulation. Moreover, the patients' circulating phagocytes displayed abnormally low levels of ROS production and markedly reduced neutrophil extracellular trap formation, altogether suggesting a role for PKCδ in activation of the NADPH oxidase complex. Our findings thus show that patients with PKCδ deficiency have impaired NADPH oxidase activity in various myeloid subsets, which may contribute to their CGD-like infectious phenotype.


Subject(s)
Infections/genetics , Protein Kinase C-delta/genetics , Respiratory Burst/physiology , B-Lymphocytes/enzymology , Female , Humans , Infant , Infections/drug therapy , Infections/etiology , Infections/pathology , Male , NADPH Oxidases/metabolism , Pedigree , Phagocytosis , Phosphorylation , Protein Isoforms , Protein Kinase C-delta/deficiency , Protein Kinase C-delta/metabolism
4.
Stem Cell Res ; 43: 101713, 2020 03.
Article in English | MEDLINE | ID: mdl-32000109

ABSTRACT

Chronic mucocutaneous candidiasis (CMC) is a disease that is characterized by susceptibility to chronic or recurrent infections with Candida spp. due to mutations affecting mainly the IL-17 signaling of T-Cells. The most common etiologies of CMC are gain-of-function (GOF) mutations in the STAT1 gene. In this paper we report the generation of a hiPSC line from a patient suffering from CMC due to a heterozygous GOF STAT1 p.R274Q mutation which can be used for disease modeling purposes.


Subject(s)
Candidiasis, Chronic Mucocutaneous/genetics , Gain of Function Mutation/genetics , Induced Pluripotent Stem Cells/metabolism , STAT1 Transcription Factor/genetics , Adolescent , Animals , Humans , Male
5.
Mol Ther Methods Clin Dev ; 17: 785-795, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32355867

ABSTRACT

Autosomal recessive (AR) complete interferon-γ receptor 1 (IFN-γR1) deficiency, also known as one genetic etiology of Mendelian susceptibility to mycobacterial disease (MSMD), is a life-threatening congenital disease leading to premature death. Affected patients present a pathognomonic predisposition to recurrent and severe infections with environmental mycobacteria or the Mycobacterium bovis bacillus Calmette-Guérin (BCG) vaccine. Current therapeutic options are limited to antibiotic treatment and hematopoietic stem cell transplantation, however with poor outcome. Given the clinical success of gene therapy, we introduce the first lentiviral-based gene therapy approach to restore expression and function of the human IFN-γR-downstream signaling cascade. In our study, we developed lentiviral vectors constitutively expressing the human IFN-γR1 and demonstrate stable transgene expression without interference with cell viability and proliferation in transduced human hematopoietic cells. Using an IFN-γR1-deficient HeLa cell model, we show stable receptor reconstitution and restored IFN-γR1 signaling without adverse effect on cell functionality. Transduction of both SV40-immortalized and primary fibroblasts derived from IFN-γR1-deficient MSMD patients was able to recover IFN-γR1 expression and restore type II IFN signaling upon stimulation with IFN-γ. In summary, we highlight lentiviral vectors to correct the IFN-γ mediated immunity and present the first gene therapy approach for patients suffering from AR complete IFN-γR1 deficiency.

6.
Cells ; 9(2)2020 02 19.
Article in English | MEDLINE | ID: mdl-32093117

ABSTRACT

Interferon γ (IFN-γ) was shown to be a macrophage activating factor already in 1984. Consistently, inborn errors of IFN-γ immunity underlie Mendelian Susceptibility to Mycobacterial Disease (MSMD). MSMD is characterized by genetic predisposition to disease caused by weakly virulent mycobacterial species. Paradoxically, macrophages from patients with MSMD were little tested. Here, we report a disease modeling platform for studying IFN-γ related pathologies using macrophages derived from patient specific induced pluripotent stem cells (iPSCs). We used iPSCs from patients with autosomal recessive complete- and partial IFN-γR2 deficiency, partial IFN-γR1 deficiency and complete STAT1 deficiency. Macrophages from all patient iPSCs showed normal morphology and IFN-γ-independent functionality like phagocytic uptake of bioparticles and internalization of cytokines. For the IFN-γ-dependent functionalities, we observed that the deficiencies played out at various stages of the IFN-γ pathway, with the complete IFN-γR2 and complete STAT1 deficient cells showing the most severe phenotypes, in terms of upregulation of surface markers and induction of downstream targets. Although iPSC-derived macrophages with partial IFN-γR1 and IFN-γR2 deficiency still showed residual induction of downstream targets, they did not reduce the mycobacterial growth when challenged with Bacillus Calmette-Guérin. Taken together, we report a disease modeling platform to study the role of macrophages in patients with inborn errors of IFN-γ immunity.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Interferon-gamma/metabolism , Macrophages/metabolism , Mycobacterium Infections/genetics , Mycobacterium , Receptors, Interferon/genetics , Blood Donors , Cellular Reprogramming , Genetic Predisposition to Disease , Humans , Mutation , Mycobacterium Infections/microbiology , Phenotype , Receptors, Interferon/deficiency , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics , Signal Transduction/genetics , Interferon gamma Receptor
7.
Stem Cell Reports ; 13(6): 957-959, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31951563

ABSTRACT

Infections with Mycobacterium tuberculosis (Mtb) are still among the top 10 causes of death worldwide, highlighting the utmost need for new forms of medical treatments. In this issue of Stem Cell Reports, Han et al. (2019) describe a technique to screen therapeutically active compounds targeting Mtb using pluripotent stem cell-derived macrophages.


Subject(s)
Antitubercular Agents/pharmacology , Drug Discovery , Macrophages/cytology , Macrophages/microbiology , Mycobacterium tuberculosis/drug effects , Pluripotent Stem Cells/cytology , Cell Differentiation , Drug Discovery/methods , Humans , Induced Pluripotent Stem Cells , Macrophages/drug effects , Macrophages/metabolism , Mycobacterium tuberculosis/physiology , Pluripotent Stem Cells/metabolism
8.
Stem Cells Transl Med ; 8(4): 332-339, 2019 04.
Article in English | MEDLINE | ID: mdl-30585439

ABSTRACT

Since their discovery in 2006, induced pluripotent stem cells (iPSCs) have opened up a world of possibilities for regenerative medicine and novel cell-based therapeutics. Now, over a decade later, robust reprogramming and expansion and differentiation protocols have been developed, and iPSC-derived cells have been used in a wide variety of small and large animal models to treat many different diseases. Furthermore, the first iPSC derivatives are on their way into clinical trials. In this line, (i) GMP-compliant generation, cultivation, and differentiation, (ii) preclinical efficacy and safety, as well as (iii) ethical and regulatory compliance of stem cell research represent important aspects that need to be evaluated for proper clinical translation of iPSCs and their derivatives. In this review article, we provide an overview of the current advances and challenges of the clinical translation of iPSC-derived blood cells and highlight the most pressing problems that have to be overcome in the next years. Stem Cells Translational Medicine 2019;8:332-339.


Subject(s)
Blood Cells/cytology , Induced Pluripotent Stem Cells/cytology , Animals , Cell Differentiation/physiology , Humans , Regenerative Medicine/methods
9.
Sci Rep ; 8(1): 16281, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30389997

ABSTRACT

Macrophages are key cells of the innate immune system and act as tissue resident macrophages (TRMs) in the homeostasis of various tissues. Given their unique functions and therapeutic use as well as the feasibility to derive macrophages in vitro from hematopoietic stem cell (HSC) sources, we propose an "easy-to-use" immune cell spray (ICS) formulation to effectively deliver HSC-derived macrophages. To achieve this aim, we used classical pump spray devices to spray either the human myeloid cell line U937 or primary murine HSC-derived macrophages. For both cell types used, one puff could deliver cells with maintained morphology and functionality. Of note, cells tolerated the spraying process very well with a recovery of more than 90%. In addition, we used osmotic preconditioning to reduce the overall cell size of macrophages. While a 800 mosm hyperosmolar sucrose solution was able to reduce the cell size by 27%, we identified 600 mosm to be effective to reduce the cell size by 15% while maintaining macrophage morphology and functionality. Using an isolated perfused rat lung preparation, the combinatorial use of the ICS with preconditioned and genetically labeled U937 cells allowed the intra-pulmonary delivery of cells, thus paving the way for a new cell delivery platform.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Macrophages/transplantation , Monocytes/transplantation , Animals , Cell Differentiation , Cell- and Tissue-Based Therapy/instrumentation , Feasibility Studies , Hematopoietic Stem Cells/physiology , Humans , K562 Cells , Lung , Macrophages/physiology , Mice , Monocytes/physiology , Osmosis , Perfusion , Primary Cell Culture/methods , Rats , U937 Cells
10.
Nat Commun ; 9(1): 5088, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504915

ABSTRACT

The increasing number of severe infections with multi-drug-resistant pathogens worldwide highlights the need for alternative treatment options. Given the pivotal role of phagocytes and especially alveolar macrophages in pulmonary immunity, we introduce a new, cell-based treatment strategy to target bacterial airway infections. Here we show that the mass production of therapeutic phagocytes from induced pluripotent stem cells (iPSC) in industry-compatible, stirred-tank bioreactors is feasible. Bioreactor-derived iPSC-macrophages (iPSC-Mac) represent a highly pure population of CD45+CD11b+CD14+CD163+ cells, and share important phenotypic, functional and transcriptional hallmarks with professional phagocytes, however with a distinct transcriptome signature similar to primitive macrophages. Most importantly, bioreactor-derived iPSC-Mac rescue mice from Pseudomonas aeruginosa-mediated acute infections of the lower respiratory tract within 4-8 h post intra-pulmonary transplantation and reduce bacterial load. Generation of specific immune-cells from iPSC-sources in scalable stirred-tank bioreactors can extend the field of immunotherapy towards bacterial infections, and may allow for further innovative cell-based treatment strategies.


Subject(s)
Bacterial Infections/prevention & control , Bioreactors , Immunotherapy/methods , Induced Pluripotent Stem Cells/cytology , Macrophages/cytology , Respiratory Tract Infections/prevention & control , Animals , Bacterial Infections/immunology , Cell Culture Techniques , Humans , Macrophages/physiology , Mice , Microscopy, Electron, Scanning , Pseudomonas aeruginosa/pathogenicity , Respiratory Tract Infections/immunology
11.
Stem Cell Reports ; 10(1): 7-16, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29249666

ABSTRACT

Mendelian susceptibility to mycobacterial disease (MSMD) is caused by inborn errors of interferon gamma (IFNγ) immunity and is characterized by severe infections by weakly virulent mycobacteria. Although IFNγ is the macrophage-activating factor, macrophages from these patients have never been studied. We demonstrate the generation of heterozygous and compound heterozygous (iMSMD-cohet) induced pluripotent stem cells (iPSCs) from a single chimeric patient, who suffered from complete autosomal recessive IFNγR1 deficiency and received bone-marrow transplantation. Loss of IFNγR1 expression had no influence on the macrophage differentiation potential of patient-specific iPSCs. In contrast, lack of IFNγR1 in iMSMD-cohet macrophages abolished IFNγ-dependent phosphorylation of STAT1 and induction of IFNγ-downstream targets such as IRF-1, SOCS-3, and IDO. As a consequence, iMSMD-cohet macrophages show impaired upregulation of HLA-DR and reduced intracellular killing of Bacillus Calmette-Guérin. We provide a disease-modeling platform that might be suited to investigate novel treatment options for MSMD and to gain insights into IFNγ signaling in macrophages.


Subject(s)
Induced Pluripotent Stem Cells/immunology , Interferon-gamma/immunology , Macrophages/immunology , Mycobacterium bovis/immunology , Receptors, Interferon/deficiency , Signal Transduction/immunology , Genetic Predisposition to Disease , Humans , Induced Pluripotent Stem Cells/microbiology , Induced Pluripotent Stem Cells/pathology , Interferon-gamma/genetics , Macrophages/microbiology , Macrophages/pathology , Receptors, Interferon/immunology , Signal Transduction/genetics , Interferon gamma Receptor
SELECTION OF CITATIONS
SEARCH DETAIL