Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Leukemia ; 38(4): 741-750, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38228679

ABSTRACT

Inflammation in the bone marrow (BM) microenvironment is a constitutive component of leukemogenesis in acute myeloid leukemia (AML). Current evidence suggests that both leukemic blasts and stroma secrete proinflammatory factors that actively suppress the function of healthy hematopoietic stem and progenitor cells (HSPCs). HSPCs are also cellular components of the innate immune system, and we reasoned that they may actively propagate the inflammation in the leukemic niche. In two separate congenic models of AML we confirm by evaluation of the BM plasma secretome and HSPC-selective single-cell RNA sequencing (scRNA-Seq) that multipotent progenitors and long-lived stem cells adopt inflammatory gene expression programs, even at low leukemic infiltration of the BM. In particular, we observe interferon gamma (IFN-γ) pathway activation, along with secretion of its chemokine target, CXCL10. We show that AML-derived nanometer-sized extracellular vesicles (EVAML) are sufficient to trigger this inflammatory HSPC response, both in vitro and in vivo. Altogether, our studies indicate that HSPCs are an unrecognized component of the inflammatory adaptation of the BM by leukemic cells. The pro-inflammatory conversion and long-lived presence of HSPCs in the BM along with their regenerative re-expansion during remission may impact clonal selection and disease evolution.


Subject(s)
Extracellular Vesicles , Leukemia, Myeloid, Acute , Humans , Hematopoietic Stem Cells/metabolism , Bone Marrow/metabolism , Leukemia, Myeloid, Acute/genetics , Inflammation/metabolism , Extracellular Vesicles/metabolism , Tumor Microenvironment
2.
Blood Adv ; 6(1): 108-120, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34625796

ABSTRACT

Bone marrow (BM) niche-derived signals are critical for facilitating engraftment after hematopoietic stem cell (HSC) transplantation (HSCT). HSCT is required for restoration of hematopoiesis in patients with inherited BM failure syndromes (iBMFSs). Shwachman-Diamond syndrome (SDS) is a rare iBMFS associated with mutations in SBDS. Previous studies have demonstrated that SBDS deficiency in osteolineage niche cells causes BM dysfunction that promotes leukemia development. However, it is unknown whether BM niche defects caused by SBDS deficiency also impair efficient engraftment of healthy donor HSC after HSCT, a hypothesis that could explain morbidity noted after clinical HSCT for patients with SDS. Here, we report a mouse model with inducible Sbds deletion in hematopoietic and osteolineage cells. Primary and secondary BM transplantation (BMT) studies demonstrated that SBDS deficiency within BM niches caused poor donor hematopoietic recovery and specifically poor HSC engraftment after myeloablative BMT. We have also identified multiple molecular and cellular defects within niche populations that are driven by SBDS deficiency and are accentuated by or develop specifically after myeloablative conditioning. These abnormalities include altered frequencies of multiple niche cell subsets, including mesenchymal lineage cells, macrophages, and endothelial cells; disruption of growth factor signaling, chemokine pathway activation, and adhesion molecule expression; and p53 pathway activation and signals involved in cell cycle arrest. Taken together, this study demonstrates that SBDS deficiency profoundly impacts recipient hematopoietic niche function in the setting of HSCT, suggesting that novel therapeutic strategies targeting host niches could improve clinical HSCT outcomes for patients with SDS.


Subject(s)
Bone Marrow , Hematopoietic Stem Cell Transplantation , Proteins , Shwachman-Diamond Syndrome , Animals , Bone Marrow/metabolism , Endothelial Cells , Gene Deletion , Hematopoiesis/genetics , Humans , Mice , Proteins/genetics , Proteins/metabolism , Shwachman-Diamond Syndrome/genetics , Shwachman-Diamond Syndrome/surgery , Transplantation Conditioning
6.
PLoS One ; 10(5): e0127414, 2015.
Article in English | MEDLINE | ID: mdl-25992652

ABSTRACT

Dyskeratosis congenita (DC) is an inherited bone marrow failure syndrome characterized by the presence of short telomeres at presentation. Mutations in ten different genes, whose products are involved in the telomere maintenance pathway, have been shown to cause DC. The X-linked form is the most common form of the disease and is caused by mutations in the gene DKC1, encoding the protein dyskerin. Dyskerin is required for the assembly and stability of telomerase and is also involved in ribosomal RNA (rRNA) processing where it converts specific uridines to pseudouridine. DC is thought to result from failure to maintain tissues, like blood, that are renewed by stem cell activity, but research into pathogenic mechanisms has been hampered by the difficulty of obtaining stem cells from patients. We reasoned that induced pluripotent stem (iPS) cells from X-linked DC patients may provide information about the mechanisms involved. Here we describe the production of iPS cells from DC patients with DKC1 mutations Q31E, A353V and ΔL37. In addition we constructed "corrected" lines with a copy of the wild type dyskerin cDNA expressed from the AAVS1 safe harbor locus. We show that in iPS cells with DKC1 mutations telomere maintenance is compromised with short telomere lengths and decreased telomerase activity. The degree to which telomere lengths are affected by expression of telomerase during reprograming, or with ectopic expression of wild type dyskerin, is variable. The recurrent mutation A353V shows the most severe effect on telomere maintenance. A353V cells but not Q31E or ΔL37 cells, are refractory to correction by expression of wild type DKC1 cDNA. Because dyskerin is involved in both telomere maintenance and ribosome biogenesis it has been postulated that defective ribosome biogenesis and translation may contribute to the disease phenotype. Evidence from mouse and zebra fish models has supported the involvement of ribosome biogenesis but primary cells from human patients have so far not shown defects in pseudouridylation or ribosomal RNA processing. None of the mutant iPS cells presented here show decreased pseudouridine levels in rRNA or defective rRNA processing suggesting telomere maintenance defects account for most of the phenotype of X-linked DC. Finally gene expression analysis of the iPS cells shows that WNT signaling is significantly decreased in all mutant cells, raising the possibility that defective WNT signaling may contribute to disease pathogenesis.


Subject(s)
Cell Cycle Proteins/genetics , Dyskeratosis Congenita/pathology , Induced Pluripotent Stem Cells/pathology , Nuclear Proteins/genetics , Ribosomes/metabolism , Telomere/pathology , Animals , Cells, Cultured , Dyskeratosis Congenita/genetics , Dyskeratosis Congenita/metabolism , Female , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mutation , Telomerase/metabolism , Wnt Signaling Pathway
7.
FEBS Lett ; 587(14): 2112-7, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23726835

ABSTRACT

Pseudouridine is the most abundant modified nucleotide in ribosomal RNA throughout eukaryotes and archaea but its role is not known. Here we produced mouse embryonic fibroblast cells expressing only catalytically inactive dyskerin, the pseudouridine synthase that converts uridine to pseudouridine in ribosomal RNA. The mutant dyskerin protein, D125A, was extremely unstable but cells were able to divide and grow very slowly. Abnormalities in ribosome RNA synthesis were apparent but mature cytoplasmic RNAs lacking pseudouridine were produced and were very unstable. We conclude that pseudouridine is required to stabilize the secondary structure of ribosomal RNA that is essential for its function.


Subject(s)
Cell Cycle Proteins/genetics , Fibroblasts/enzymology , Nuclear Proteins/genetics , Pseudouridine/metabolism , RNA, Ribosomal/metabolism , Amino Acid Substitution , Animals , Cell Cycle Proteins/metabolism , Cell Proliferation , Cells, Cultured , Embryo, Mammalian/cytology , Enzyme Stability , Fibroblasts/physiology , Mice , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nuclear Proteins/metabolism , RNA Processing, Post-Transcriptional , RNA Stability , RNA, Small Nucleolar/metabolism
8.
Aging Cell ; 10(2): 338-48, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21241452

ABSTRACT

Mutations in DKC1, encoding telomerase associated protein dyskerin, cause X-linked dyskeratosis congenita (DC), a bone marrow (BM) failure, and cancer susceptibility syndrome. Decreased accumulation of telomerase RNA resulting in excessive telomere shortening and premature cellular senescence is thought to be the primary cause of disease in X-linked DC. Affected tissues are those that require constant renewal by stem cell activity. We previously showed that in Dkc1(Δ15) mice, which contain a mutation that is a copy of a human mutation causing DC, mutant cells have a telomerase-dependent proliferative defect and increased accumulation of DNA damage in the first generation before the telomeres are short. We now demonstrate the presence of the growth defect in Dkc1(Δ15) mouse embryonic fibroblasts in vitro and show that accumulation of DNA damage and levels of reactive oxygen species increase with increasing population doublings. Treatment with the antioxidant, N-acetyl cysteine (NAC), partially rescued the growth disadvantage of mutant cells in vitro and in vivo. Competitive BM repopulation experiments showed that the Dkc1(Δ15) mutation is associated with a functional stem cell defect that becomes more severe with increasing age, consistent with accelerated senescence, a hallmark of DC hematopoiesis. This stem cell phenotype was partially corrected by NAC treatment. These results suggest that a pathogenic Dkc1 mutation accelerates stem cell aging, that increased oxidative stress might play a role in the pathogenesis of X-linked DC, and that some manifestations of DC may be prevented or delayed by antioxidant treatment.


Subject(s)
Antioxidants/therapeutic use , Cell Cycle Proteins/metabolism , Cellular Senescence/physiology , Dyskeratosis Congenita/drug therapy , Dyskeratosis Congenita/physiopathology , Hematopoietic Stem Cells/physiology , Nuclear Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Dyskeratosis Congenita/pathology , Female , Fibroblasts/cytology , Fibroblasts/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Telomerase/genetics , Telomerase/metabolism , Telomere/metabolism , Telomere/pathology
9.
FEBS Lett ; 583(18): 3086-90, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19729012

ABSTRACT

Mutations in the X-linked gene, DKC1, encoding dyskerin, cause dyskeratosis congenita by leading to decreased telomerase activity and causing short telomeres. Dyskerin is also a pseudouridine synthase that modifies nascent ribosomal and other RNAs and it is not known if this function is affected by the mutations. Here we show that newly synthesized ribosomal RNA, extracted from human and mouse cells with pathogenic mutations, shows anomalous mobility in agarose gels under certain denaturation conditions. The anomalously migrating RNA is turned over rapidly. Analysis of ribosomal RNA in these cells suggests the altered mobility is due to inefficient pseudouridylation.


Subject(s)
Cell Cycle Proteins/genetics , Electrophoresis , Mutation , Nuclear Proteins/genetics , RNA, Ribosomal/chemistry , Animals , Humans , Mice , Pseudouridine/metabolism , RNA Stability , RNA, Ribosomal/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...