Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
Clin Immunol ; 256: 109777, 2023 11.
Article in English | MEDLINE | ID: mdl-37741518

ABSTRACT

C-terminal variants in CDC42 encoding cell division control protein 42 homolog underlie neonatal-onset cytopenia, autoinflammation, rash, and hemophagocytic lymphohistiocytosis (NOCARH). Pyrin inflammasome hyperactivation has been shown to contribute to disease pathophysiology. However, mortality of NOCARH patients remains high despite inflammasome-focused treatments. Here, we demonstrate in four NOCARH patients from three families that cell-intrinsic activation of type I interferon (IFN) is a previously unrecognized driver of autoinflammation in NOCARH. Our data show that aberrant innate immune activation is caused by sensing of cytosolic nucleic acids released from mitochondria, which exhibit disturbances in integrity and dynamics due to CDC42 dysfunction. In one of our patients, treatment with the Janus kinase inhibitor ruxolitinib led to complete remission, indicating that inhibition of type I IFN signaling may have an important role in the management of autoinflammation in patients with NOCARH.


Subject(s)
Interferon Type I , Lymphohistiocytosis, Hemophagocytic , Humans , Infant, Newborn , cdc42 GTP-Binding Protein , Inflammasomes/genetics , Lymphohistiocytosis, Hemophagocytic/etiology , Nitriles , Syndrome
2.
Exp Hematol ; 133: 104207, 2024 May.
Article in English | MEDLINE | ID: mdl-38522505

ABSTRACT

Myelodysplastic/myeloproliferative diseases of childhood cause a relevant disease burden, and many of these diseases may have a fatal course. The use of next-generation sequencing (NGS) has led to the identification of novel genetic variants in patients with these diseases, advancing our understanding of the underlying pathophysiology. However, novel mutations can often only be interpreted as variants of unknown significance (VUS), hindering adequate diagnosis and the use of a targeted therapy. To improve variant interpretation and test targeted therapies in a preclinical setting, we are using a rapid zebrafish embryo model that allows functional evaluation of the novel variant and possible therapeutic approaches within days. Thereby, we accelerate the translation from genetic findings to treatment options. Here, we establish this workflow on a novel in-frame tandem duplication in NRAS (c.192_227dup; p.G75_E76insDS65_G75) identified by Sanger sequencing in a 2.5-year-old patient with an unclassifiable myelodysplastic/myeloproliferative neoplasm (MDS/MPN-U). We show that this variant results in a myeloproliferative phenotype in zebrafish embryos with expansion of immature myeloid cells in the caudal hematopoietic tissue, which can be reversed by MEK inhibition. Thus, we could reclassify the variant from likely pathogenic to pathogenic using the American College of Medical Genetics (ACMG) criteria.


Subject(s)
GTP Phosphohydrolases , Membrane Proteins , Myelodysplastic-Myeloproliferative Diseases , Zebrafish , Animals , Child, Preschool , Female , Humans , Male , Gene Duplication , GTP Phosphohydrolases/genetics , High-Throughput Nucleotide Sequencing , Membrane Proteins/genetics , Myelodysplastic-Myeloproliferative Diseases/genetics , Tandem Repeat Sequences , Zebrafish/genetics
3.
Nat Commun ; 9(1): 3090, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30082823

ABSTRACT

The H2.0-like homeobox transcription factor (HLX) regulates hematopoietic differentiation and is overexpressed in Acute Myeloid Leukemia (AML), but the mechanisms underlying these functions remain unclear. We demonstrate here that HLX overexpression leads to a myeloid differentiation block both in zebrafish and human hematopoietic stem and progenitor cells (HSPCs). We show that HLX overexpression leads to downregulation of genes encoding electron transport chain (ETC) components and upregulation of PPARδ gene expression in zebrafish and human HSPCs. HLX overexpression also results in AMPK activation. Pharmacological modulation of PPARδ signaling relieves the HLX-induced myeloid differentiation block and rescues HSPC loss upon HLX knockdown but it has no effect on AML cell lines. In contrast, AMPK inhibition results in reduced viability of AML cell lines, but minimally affects myeloid progenitors. This newly described role of HLX in regulating the metabolic state of hematopoietic cells may have important therapeutic implications.


Subject(s)
Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/physiology , Leukemia, Myeloid, Acute/metabolism , Transcription Factors/physiology , Zebrafish Proteins/physiology , Animals , Autophagy , Cell Differentiation , Cell Proliferation , Cell Survival , Gene Expression Regulation, Leukemic , Hematopoiesis , Homeodomain Proteins/genetics , Humans , K562 Cells , Leukemia, Myeloid, Acute/genetics , Membrane Potential, Mitochondrial , PPAR gamma/metabolism , Phenotype , Reactive Oxygen Species/metabolism , Signal Transduction , Stem Cells/metabolism , Transcription Factors/genetics , Zebrafish , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL