Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 22(11): 1382-1390, 2021 11.
Article in English | MEDLINE | ID: mdl-34663978

ABSTRACT

Intergenerational inheritance of immune traits linked to epigenetic modifications has been demonstrated in plants and invertebrates. Here we provide evidence for transmission of trained immunity across generations to murine progeny that survived a sublethal systemic infection with Candida albicans or a zymosan challenge. The progeny of trained mice exhibited cellular, developmental, transcriptional and epigenetic changes associated with the bone marrow-resident myeloid effector and progenitor cell compartment. Moreover, the progeny of trained mice showed enhanced responsiveness to endotoxin challenge, alongside improved protection against systemic heterologous Escherichia coli and Listeria monocytogenes infections. Sperm DNA of parental male mice intravenously infected with the fungus C. albicans showed DNA methylation differences linked to immune gene loci. These results provide evidence for inheritance of trained immunity in mammals, enhancing protection against infections.


Subject(s)
Candida albicans/immunology , Candidiasis/immunology , Escherichia coli Infections/immunology , Escherichia coli/immunology , Heredity , Immunity, Innate/genetics , Listeria monocytogenes/immunology , Listeriosis/immunology , Myeloid Cells/immunology , Animals , Candida albicans/pathogenicity , Candidiasis/genetics , Candidiasis/metabolism , Candidiasis/microbiology , Cells, Cultured , DNA Methylation , Disease Models, Animal , Epigenesis, Genetic , Escherichia coli/pathogenicity , Escherichia coli Infections/genetics , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Host-Pathogen Interactions , Listeria monocytogenes/pathogenicity , Listeriosis/genetics , Listeriosis/metabolism , Listeriosis/microbiology , Male , Mice, Transgenic , Myeloid Cells/metabolism , Myeloid Cells/microbiology , Spermatozoa/immunology , Spermatozoa/metabolism , Transcription, Genetic
2.
Nat Immunol ; 21(1): 75-85, 2020 01.
Article in English | MEDLINE | ID: mdl-31844326

ABSTRACT

Regulatory T (Treg) cells accumulate into tumors, hindering the success of cancer immunotherapy. Yet, therapeutic targeting of Treg cells shows limited efficacy or leads to autoimmunity. The molecular mechanisms that guide Treg cell stability in tumors remain elusive. In the present study, we identify a cell-intrinsic role of the alarmin interleukin (IL)-33 in the functional stability of Treg cells. Specifically, IL-33-deficient Treg cells demonstrated attenuated suppressive properties in vivo and facilitated tumor regression in a suppression of tumorigenicity 2 receptor (ST2) (IL-33 receptor)-independent fashion. On activation, Il33-/- Treg cells exhibited epigenetic re-programming with increased chromatin accessibility of the Ifng locus, leading to elevated interferon (IFN)-γ production in a nuclear factor (NF)-κB-T-bet-dependent manner. IFN-γ was essential for Treg cell defective function because its ablation restored Il33-/- Treg cell-suppressive properties. Importantly, genetic ablation of Il33 potentiated the therapeutic effect of immunotherapy. Our findings reveal a new and therapeutically important intrinsic role of IL-33 in Treg cell stability in cancer.


Subject(s)
Interferon-gamma/immunology , Interleukin-33/immunology , Melanoma, Experimental/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Escape/immunology , Animals , Cell Line, Tumor , Interferon-gamma/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism
4.
Immunity ; 54(11): 2650-2669.e14, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34592166

ABSTRACT

Longitudinal analyses of the innate immune system, including the earliest time points, are essential to understand the immunopathogenesis and clinical course of coronavirus disease (COVID-19). Here, we performed a detailed characterization of natural killer (NK) cells in 205 patients (403 samples; days 2 to 41 after symptom onset) from four independent cohorts using single-cell transcriptomics and proteomics together with functional studies. We found elevated interferon (IFN)-α plasma levels in early severe COVD-19 alongside increased NK cell expression of IFN-stimulated genes (ISGs) and genes involved in IFN-α signaling, while upregulation of tumor necrosis factor (TNF)-induced genes was observed in moderate diseases. NK cells exert anti-SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) activity but are functionally impaired in severe COVID-19. Further, NK cell dysfunction may be relevant for the development of fibrotic lung disease in severe COVID-19, as NK cells exhibited impaired anti-fibrotic activity. Our study indicates preferential IFN-α and TNF responses in severe and moderate COVID-19, respectively, and associates a prolonged IFN-α-induced NK cell response with poorer disease outcome.


Subject(s)
COVID-19/immunology , Interferon-alpha/immunology , Killer Cells, Natural/immunology , SARS-CoV-2/immunology , Tumor Necrosis Factor-alpha/metabolism , Base Sequence , Humans , Immunity, Innate/immunology , Inflammation/immunology , Interferon-alpha/blood , Pulmonary Fibrosis/pathology , RNA-Seq , Severity of Illness Index , Transcriptome/genetics , United Kingdom , United States
6.
Nature ; 594(7862): 265-270, 2021 06.
Article in English | MEDLINE | ID: mdl-34040261

ABSTRACT

Fast and reliable detection of patients with severe and heterogeneous illnesses is a major goal of precision medicine1,2. Patients with leukaemia can be identified using machine learning on the basis of their blood transcriptomes3. However, there is an increasing divide between what is technically possible and what is allowed, because of privacy legislation4,5. Here, to facilitate the integration of any medical data from any data owner worldwide without violating privacy laws, we introduce Swarm Learning-a decentralized machine-learning approach that unites edge computing, blockchain-based peer-to-peer networking and coordination while maintaining confidentiality without the need for a central coordinator, thereby going beyond federated learning. To illustrate the feasibility of using Swarm Learning to develop disease classifiers using distributed data, we chose four use cases of heterogeneous diseases (COVID-19, tuberculosis, leukaemia and lung pathologies). With more than 16,400 blood transcriptomes derived from 127 clinical studies with non-uniform distributions of cases and controls and substantial study biases, as well as more than 95,000 chest X-ray images, we show that Swarm Learning classifiers outperform those developed at individual sites. In addition, Swarm Learning completely fulfils local confidentiality regulations by design. We believe that this approach will notably accelerate the introduction of precision medicine.


Subject(s)
Blockchain , Clinical Decision-Making/methods , Confidentiality , Datasets as Topic , Machine Learning , Precision Medicine/methods , COVID-19/diagnosis , COVID-19/epidemiology , Disease Outbreaks , Female , Humans , Leukemia/diagnosis , Leukemia/pathology , Leukocytes/pathology , Lung Diseases/diagnosis , Machine Learning/trends , Male , Software , Tuberculosis/diagnosis
7.
PLoS Genet ; 20(3): e1011142, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38457455

ABSTRACT

Succinate is a potent immune signalling molecule that is present in the mammalian gut and within macrophages. Both of these infection niches are colonised by the pathogenic bacterium Salmonella enterica serovar Typhimurium during infection. Succinate is a C4-dicarboyxlate that can serve as a source of carbon for bacteria. When succinate is provided as the sole carbon source for in vitro cultivation, Salmonella and other enteric bacteria exhibit a slow growth rate and a long lag phase. This growth inhibition phenomenon was known to involve the sigma factor RpoS, but the genetic basis of the repression of bacterial succinate utilisation was poorly understood. Here, we use an experimental evolution approach to isolate fast-growing mutants during growth of S. Typhimurium on succinate containing minimal medium. Our approach reveals novel RpoS-independent systems that inhibit succinate utilisation. The CspC RNA binding protein restricts succinate utilisation, an inhibition that is antagonised by high levels of the small regulatory RNA (sRNA) OxyS. We discovered that the Fe-S cluster regulatory protein IscR inhibits succinate utilisation by repressing the C4-dicarboyxlate transporter DctA. Furthermore, the ribose operon repressor RbsR is required for the complete RpoS-driven repression of succinate utilisation, suggesting a novel mechanism of RpoS regulation. Our discoveries shed light on the redundant regulatory systems that tightly regulate the utilisation of succinate. We speculate that the control of central carbon metabolism by multiple regulatory systems in Salmonella governs the infection niche-specific utilisation of succinate.


Subject(s)
Bacterial Proteins , Succinic Acid , Animals , Bacterial Proteins/metabolism , Succinic Acid/metabolism , Salmonella typhimurium/genetics , Succinates/metabolism , Carbon/metabolism , Sigma Factor/genetics , Sigma Factor/metabolism , Gene Expression Regulation, Bacterial , Mammals/metabolism
8.
EMBO J ; 41(23): e110595, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36305367

ABSTRACT

Mammalian SWI/SNF/BAF chromatin remodeling complexes influence cell lineage determination. While the contribution of these complexes to neural progenitor cell (NPC) proliferation and differentiation has been reported, little is known about the transcriptional profiles that determine neurogenesis or gliogenesis. Here, we report that BCL7A is a modulator of the SWI/SNF/BAF complex that stimulates the genome-wide occupancy of the ATPase subunit BRG1. We demonstrate that BCL7A is dispensable for SWI/SNF/BAF complex integrity, whereas it is essential to regulate Notch/Wnt pathway signaling and mitochondrial bioenergetics in differentiating NPCs. Pharmacological stimulation of Wnt signaling restores mitochondrial respiration and attenuates the defective neurogenic patterns observed in NPCs lacking BCL7A. Consistently, treatment with an enhancer of mitochondrial biogenesis, pioglitazone, partially restores mitochondrial respiration and stimulates neuronal differentiation of BCL7A-deficient NPCs. Using conditional BCL7A knockout mice, we reveal that BCL7A expression in NPCs and postmitotic neurons is required for neuronal plasticity and supports behavioral and cognitive performance. Together, our findings define the specific contribution of BCL7A-containing SWI/SNF/BAF complexes to mitochondria-driven NPC commitment, thereby providing a better understanding of the cell-intrinsic transcriptional processes that connect metabolism, neuronal morphogenesis, and cognitive flexibility.


Subject(s)
Cell Differentiation , Microfilament Proteins , Neural Stem Cells , Animals , Mice , Adenosine Triphosphatases/metabolism , Chromatin Assembly and Disassembly , Energy Metabolism , Mitochondria/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Microfilament Proteins/metabolism , Neural Stem Cells/cytology
9.
Immunity ; 47(6): 1051-1066.e12, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29262348

ABSTRACT

Human in vitro generated monocyte-derived dendritic cells (moDCs) and macrophages are used clinically, e.g., to induce immunity against cancer. However, their physiological counterparts, ontogeny, transcriptional regulation, and heterogeneity remains largely unknown, hampering their clinical use. High-dimensional techniques were used to elucidate transcriptional, phenotypic, and functional differences between human in vivo and in vitro generated mononuclear phagocytes to facilitate their full potential in the clinic. We demonstrate that monocytes differentiated by macrophage colony-stimulating factor (M-CSF) or granulocyte macrophage colony-stimulating factor (GM-CSF) resembled in vivo inflammatory macrophages, while moDCs resembled in vivo inflammatory DCs. Moreover, differentiated monocytes presented with profound transcriptomic, phenotypic, and functional differences. Monocytes integrated GM-CSF and IL-4 stimulation combinatorically and temporally, resulting in a mode- and time-dependent differentiation relying on NCOR2. Finally, moDCs are phenotypically heterogeneous and therefore necessitate the use of high-dimensional phenotyping to open new possibilities for better clinical tailoring of these cellular therapies.


Subject(s)
Dendritic Cells/immunology , Interleukin-4/immunology , Macrophages/immunology , Monocytes/immunology , Nuclear Receptor Co-Repressor 2/immunology , Signal Transduction/immunology , Cell Differentiation , Cell Lineage , Dendritic Cells/cytology , Dendritic Cells/drug effects , Gene Expression Profiling , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunophenotyping , Interleukin-4/genetics , Interleukin-4/pharmacology , Macrophage Activation , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/cytology , Macrophages/drug effects , Monocytes/cytology , Monocytes/drug effects , Nuclear Receptor Co-Repressor 2/genetics , Primary Cell Culture , Time Factors , Transcription, Genetic
10.
EMBO J ; 40(24): e108662, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34825707

ABSTRACT

Chronic neuroinflammation is a pathogenic component of Alzheimer's disease (AD) that may limit the ability of the brain to clear amyloid deposits and cellular debris. Tight control of the immune system is therefore key to sustain the ability of the brain to repair itself during homeostasis and disease. The immune-cell checkpoint receptor/ligand pair PD-1/PD-L1, known for their inhibitory immune function, is expressed also in the brain. Here, we report upregulated expression of PD-L1 and PD-1 in astrocytes and microglia, respectively, surrounding amyloid plaques in AD patients and in the APP/PS1 AD mouse model. We observed juxtamembrane shedding of PD-L1 from astrocytes, which may mediate ectodomain signaling to PD-1-expressing microglia. Deletion of microglial PD-1 evoked an inflammatory response and compromised amyloid-ß peptide (Aß) uptake. APP/PS1 mice deficient for PD-1 exhibited increased deposition of Aß, reduced microglial Aß uptake, and decreased expression of the Aß receptor CD36 on microglia. Therefore, ineffective immune regulation by the PD-1/PD-L1 axis contributes to Aß plaque deposition during chronic neuroinflammation in AD.


Subject(s)
Alzheimer Disease/immunology , Amyloid beta-Protein Precursor/genetics , B7-H1 Antigen/metabolism , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Up-Regulation , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/toxicity , Animals , Astrocytes/metabolism , CD36 Antigens/metabolism , Case-Control Studies , Disease Models, Animal , Female , Gene Deletion , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Mice, Transgenic , Microglia/metabolism , Middle Aged
11.
EMBO J ; 40(18): e107100, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34337766

ABSTRACT

Adult neurogenesis enables the life-long addition of functional neurons to the hippocampus and is regulated by both cell-intrinsic molecular programs and behavioral activity. De novo DNA methylation is crucial for embryonic brain development, but its role during adult hippocampal neurogenesis has remained unknown. Here, we show that de novo DNA methylation is critical for maturation and functional integration of adult-born neurons in the mouse hippocampus. Bisulfite sequencing revealed that de novo DNA methyltransferases target neuronal enhancers and gene bodies during adult hippocampal neural stem cell differentiation, to establish neuronal methylomes and facilitate transcriptional up-regulation of neuronal genes. Inducible deletion of both de novo DNA methyltransferases Dnmt3a and Dnmt3b in adult neural stem cells did not affect proliferation or fate specification, but specifically impaired dendritic outgrowth and synaptogenesis of newborn neurons, thereby hampering their functional maturation. Consequently, abolishing de novo DNA methylation modulated activation patterns in the hippocampal circuitry and caused specific deficits in hippocampus-dependent learning and memory. Our results demonstrate that proper establishment of neuronal methylomes during adult neurogenesis is fundamental for hippocampal function.


Subject(s)
Cell Differentiation/genetics , DNA Methylation , Hippocampus/physiology , Neurogenesis/genetics , Pyramidal Cells/cytology , Pyramidal Cells/metabolism , Animals , Cells, Cultured , Epigenesis, Genetic , Gene Expression Regulation , Mice
12.
EMBO Rep ; 23(8): e54315, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35695071

ABSTRACT

The primary cilium constitutes an organelle that orchestrates signal transduction independently from the cell body. Dysregulation of this intricate molecular architecture leads to severe human diseases, commonly referred to as ciliopathies. However, the molecular underpinnings how ciliary signaling orchestrates a specific cellular output remain elusive. By combining spatially resolved optogenetics with RNA sequencing and imaging, we reveal a novel cAMP signalosome that is functionally distinct from the cytoplasm. We identify the genes and pathways targeted by the ciliary cAMP signalosome and shed light on the underlying mechanisms and downstream signaling. We reveal that chronic stimulation of the ciliary cAMP signalosome transforms kidney epithelia from tubules into cysts. Counteracting this chronic cAMP elevation in the cilium by small molecules targeting activation of phosphodiesterase-4 long isoforms inhibits cyst growth. Thereby, we identify a novel concept of how the primary cilium controls cellular functions and maintains tissue integrity in a specific and spatially distinct manner and reveal novel molecular components that might be involved in the development of one of the most common genetic diseases, polycystic kidney disease.


Subject(s)
Cysts , Polycystic Kidney Diseases , Cilia/metabolism , Cysts/metabolism , Gene Expression , Humans , Kidney , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism
13.
EMBO Rep ; 23(5): e52606, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35297148

ABSTRACT

Mitochondrial dysfunction can either extend or decrease Caenorhabditis elegans lifespan, depending on whether transcriptionally regulated responses can elicit durable stress adaptation to otherwise detrimental lesions. Here, we test the hypothesis that enhanced metabolic flexibility is sufficient to circumvent bioenergetic abnormalities associated with the phenotypic threshold effect, thereby transforming short-lived mitochondrial mutants into long-lived ones. We find that CEST-2.2, a carboxylesterase mainly localizes in the intestine, may stimulate the survival of mitochondrial deficient animals. We report that genetic manipulation of cest-2.2 expression has a minor lifespan impact on wild-type nematodes, whereas its overexpression markedly extends the lifespan of complex I-deficient gas-1(fc21) mutants. We profile the transcriptome and lipidome of cest-2.2 overexpressing animals and show that CEST-2.2 stimulates lipid metabolism and fatty acid beta-oxidation, thereby enhancing mitochondrial respiratory capacity through complex II and LET-721/ETFDH, despite the inherited genetic lesion of complex I. Together, our findings unveil a metabolic pathway that, through the tissue-specific mobilization of lipid deposits, may influence the longevity of mitochondrial mutant C. elegans.


Subject(s)
Caenorhabditis elegans Proteins , Longevity , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Lipid Metabolism/genetics , Longevity/genetics , Mitochondria/metabolism
14.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33372158

ABSTRACT

Macrophages are the principal immune cells of the epididymis and testis, but their origins, heterogeneity, development, and maintenance are not well understood. Here, we describe distinct populations of epididymal and testicular macrophages that display an organ-specific cellular identity. Combining in vivo fate-mapping, chimeric and parabiotic mouse models with in-depth cellular analyses, we found that CD64hiMHCIIlo and CD64loMHCIIhi macrophage populations of epididymis and testis arise sequentially from yolk sac erythro-myeloid progenitors, embryonic hematopoiesis, and nascent neonatal monocytes. While monocytes were the major developmental source of both epididymal and testicular macrophages, both populations self-maintain in the steady-state independent of bone marrow hematopoietic precursors. However, after radiation-induced macrophage ablation or during infection, bone marrow-derived circulating monocytes are recruited to the epididymis and testis, giving rise to inflammatory macrophages that promote tissue damage. These results define the layered ontogeny, maintenance and inflammatory response of macrophage populations in the male reproductive organs.


Subject(s)
Infertility, Male/immunology , Macrophages/immunology , Macrophages/metabolism , Animals , Cell Differentiation , Cell Lineage , Epididymis/immunology , Epididymis/metabolism , Infertility, Male/metabolism , Infertility, Male/physiopathology , Male , Mice , Mice, Inbred C57BL , Monocytes/immunology , Testis/immunology , Testis/metabolism
15.
Proc Natl Acad Sci U S A ; 118(31)2021 08 03.
Article in English | MEDLINE | ID: mdl-34326259

ABSTRACT

Proinflammatory activation of macrophages in metabolic tissues is critically important in the induction of obesity-induced metaflammation. Here, we demonstrate that the soluble mannose receptor (sMR) plays a direct functional role in both macrophage activation and metaflammation. We show that sMR binds CD45 on macrophages and inhibits its phosphatase activity, leading to an Src/Akt/NF-κB-mediated cellular reprogramming toward an inflammatory phenotype both in vitro and in vivo. Remarkably, increased serum sMR levels were observed in obese mice and humans and directly correlated with body weight. Importantly, enhanced sMR levels increase serum proinflammatory cytokines, activate tissue macrophages, and promote insulin resistance. Altogether, our results reveal sMR as regulator of proinflammatory macrophage activation, which could constitute a therapeutic target for metaflammation and other hyperinflammatory diseases.


Subject(s)
Gene Expression Regulation/drug effects , Macrophage Activation/drug effects , Macrophages/metabolism , Mannose Receptor/chemistry , Membrane Proteins/pharmacology , Animal Feed , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Diet, High-Fat , Gastrointestinal Microbiome , Inflammation , Macrophage Activation/physiology , Male , Mannose Receptor/metabolism , Mice , Mice, Knockout , Random Allocation
16.
RNA ; 2021 May 11.
Article in English | MEDLINE | ID: mdl-33975917

ABSTRACT

The stem cell-specific RNA-binding protein TRIM71/LIN-41 was the first identified target of the pro-differentiation and tumor suppressor miRNA let-7. TRIM71 has essential functions in embryonic development and a proposed oncogenic role in several cancer types, such as hepatocellular carcinoma. Here, we show that TRIM71 regulates let-7 expression and activity via two independent mechanisms. On the one hand, TRIM71 enhances pre-let-7 degradation through its direct interaction with LIN28 and TUT4, thereby inhibiting let-7 maturation and indirectly promoting the stabilization of let-7 targets. On the other hand, TRIM71 represses the activity of mature let-7 via its RNA-dependent interaction with the RNA-Induced Silencing Complex (RISC) effector protein AGO2. We found that TRIM71 directly binds and stabilizes let-7 targets, suggesting that let-7 activity inhibition occurs on active RISCs. MiRNA enrichment analysis of several transcriptomic datasets from mouse embryonic stem cells and human hepatocellular carcinoma cells suggests that these let-7 regulatory mechanisms shape transcriptomic changes during developmental and oncogenic processes. Altogether, our work reveals a novel role for TRIM71 as a miRNA repressor and sheds light on a dual mechanism of let-7 regulation.

17.
Int J Mol Sci ; 23(4)2022 Feb 17.
Article in English | MEDLINE | ID: mdl-35216353

ABSTRACT

X-linked dystonia-parkinsonism (XDP) is a severe neurodegenerative disorder that manifests as adult-onset dystonia combined with parkinsonism. A SINE-VNTR-Alu (SVA) retrotransposon inserted in an intron of the TAF1 gene reduces its expression and alters splicing in XDP patient-derived cells. As a consequence, increased levels of the TAF1 intron retention transcript TAF1-32i can be found in XDP cells as compared to healthy controls. Here, we investigate the sequence of the deep intronic region included in this transcript and show that it is also present in cells from healthy individuals, albeit in lower amounts than in XDP cells, and that it undergoes degradation by nonsense-mediated mRNA decay. Furthermore, we investigate epigenetic marks (e.g., DNA methylation and histone modifications) present in this intronic region and the spanning sequence. Finally, we show that the SVA evinces regulatory potential, as demonstrated by its ability to repress the TAF1 promoter in vitro. Our results enable a better understanding of the disease mechanisms underlying XDP and transcriptional alterations caused by SVA retrotransposons.


Subject(s)
Dystonic Disorders/genetics , Genetic Diseases, X-Linked/genetics , Parkinsonian Disorders/genetics , Retroelements/genetics , Transcription, Genetic/genetics , Adolescent , Adult , DNA Methylation/genetics , Female , Histone Acetyltransferases/genetics , Humans , Introns/genetics , Male , Middle Aged , Promoter Regions, Genetic/genetics , Short Interspersed Nucleotide Elements/genetics , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Young Adult
18.
J Mol Cell Cardiol ; 156: 45-56, 2021 07.
Article in English | MEDLINE | ID: mdl-33773996

ABSTRACT

CRELD1 (Cysteine-Rich with EGF-Like Domains 1) is a risk gene for non-syndromic atrioventricular septal defects in human patients. In a mouse model, Creld1 has been shown to be essential for heart development, particularly in septum and valve formation. However, due to the embryonic lethality of global Creld1 knockout (KO) mice, its cell type-specific function during peri- and postnatal stages remains unknown. Here, we generated conditional Creld1 KO mice lacking Creld1 either in the endocardium (KOTie2) or the myocardium (KOMyHC). Using a combination of cardiac phenotyping, histology, immunohistochemistry, RNA-sequencing, and flow cytometry, we demonstrate that Creld1 function in the endocardium is dispensable for heart development. Lack of myocardial Creld1 causes extracellular matrix remodeling and trabeculation defects by modulation of the Notch1 signaling pathway. Hence, KOMyHC mice die early postnatally due to myocardial hypoplasia. Our results reveal that Creld1 not only controls the formation of septa and valves at an early stage during heart development, but also cardiac maturation and function at a later stage. These findings underline the central role of Creld1 in mammalian heart development and function.


Subject(s)
Cell Adhesion Molecules/genetics , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Developmental , Heart/embryology , Heart/physiology , Myocardium/metabolism , Organogenesis/genetics , Animals , Biomarkers , Cell Adhesion Molecules/metabolism , Extracellular Matrix Proteins/metabolism , Flow Cytometry , Gene Expression Profiling , Humans , Mice, Knockout , Single-Cell Analysis
19.
Glia ; 67(4): 619-633, 2019 04.
Article in English | MEDLINE | ID: mdl-30585358

ABSTRACT

Astrocytes support normal brain function, but may also contribute to neurodegeneration when they become reactive under pathological conditions such as stroke. However, the molecular underpinnings of this context-dependent interplay between beneficial and detrimental properties in reactive astrogliosis have remained incompletely understood. Therefore, using the RiboTag technique, we immunopurified translating mRNAs specifically from astrocytes 72 hr after transient middle cerebral artery occlusion in mice (tMCAO), thereby generating a stroke-specific astroglial translatome database. We found that compared to control brains, reactive astrocytes after tMCAO show an enrichment of transcripts linked to the A2 phenotype, which has been associated with neuroprotection. However, we found that astrocytes also upregulate a large number of potentially neurotoxic genes. In total, we identified the differential expression of 1,003 genes and 38 transcription factors, of which Stat3, Sp1, and Spi1 were the most prominent. To further explore the effects of Stat3-mediated pathways on stroke pathogenesis, we subjected mice with an astrocyte-specific conditional deletion of Stat3 to tMCAO, and found that these mice have reduced stroke volume and improved motor outcome 72 hr after focal ischemia. Taken together, our study extends the emerging database of novel astrocyte-specific targets for stroke therapy, and supports the role of astrocytes as critical safeguards of brain function in health and disease.


Subject(s)
Astrocytes/metabolism , Gene Expression Profiling/methods , Infarction, Middle Cerebral Artery/pathology , Rhombencephalon/pathology , Animals , Computational Biology , Connexin 43/genetics , Connexin 43/metabolism , Disease Models, Animal , Female , Galectin 3/genetics , Galectin 3/metabolism , Gene Expression Regulation/genetics , Immunoprecipitation , Infarction, Middle Cerebral Artery/physiopathology , Lipocalin-2/genetics , Lipocalin-2/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Rotarod Performance Test , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
20.
Mol Ther ; 26(1): 95-104, 2018 01 03.
Article in English | MEDLINE | ID: mdl-29103909

ABSTRACT

The chemokine CCL17, mainly produced by dendritic cells (DCs) in the immune system, is involved in the pathogenesis of various inflammatory diseases. As a ligand of CCR4, CCL17 induces chemotaxis and facilitates T cell-DC interactions. We report the identification of two novel RNA aptamers, which were validated in vitro and in vivo for their capability to neutralize CCL17. Both aptamers efficiently inhibited the directed migration of the CCR4+ lymphoma line BW5147.3 toward CCL17 in a dose-dependent manner. To study the effect of these aptamers in vivo, we used a murine model of contact hypersensitivity. Systemic application of the aptamers significantly prevented ear swelling and T cell infiltration into the ears of sensitized mice after challenge with the contact sensitizer. The results of this proof-of-principle study establish aptamers as potent inhibitors of CCL17-mediated chemotaxis. Potentially, CCL17-specific aptamers may be used therapeutically in humans to treat or prevent allergic and inflammatory diseases.


Subject(s)
Aptamers, Nucleotide/genetics , Chemokine CCL17/genetics , Chemotaxis/genetics , Chemotaxis/immunology , Dermatitis, Contact/genetics , Dermatitis, Contact/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Aptamers, Nucleotide/chemistry , Cell Movement/genetics , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Macrophages/immunology , Macrophages/metabolism , Mice , Nucleic Acid Conformation , SELEX Aptamer Technique
SELECTION OF CITATIONS
SEARCH DETAIL