Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Cell Rep ; 34(4): 108656, 2021 01 26.
Article in English | MEDLINE | ID: mdl-33503437

ABSTRACT

Muscle satellite cells (SCs) are a quiescent (non-proliferative) stem cell population in uninjured skeletal muscle. Although SCs have been investigated for nearly 60 years, the molecular drivers that transform quiescent SCs into the rapidly dividing (activated) stem/progenitor cells that mediate muscle repair after injury remain largely unknown. Here we identify a prominent FBJ osteosarcoma oncogene (Fos) mRNA and protein signature in recently activated SCs that is rapidly, heterogeneously, and transiently induced by muscle damage. We further reveal a requirement for FOS to efficiently initiate key stem cell functions, including cell cycle entry, proliferative expansion, and muscle regeneration, via induction of "pro-regenerative" target genes that stimulate cell migration, division, and differentiation. Disruption of one of these Fos/AP-1 targets, NAD(+)-consuming mono-ADP-ribosyl-transferase 1 (Art1), in SCs delays cell cycle entry and impedes progenitor cell expansion and muscle regeneration. This work uncovers an early-activated FOS/ART1/mono-ADP-ribosylation (MARylation) pathway that is essential for stem cell-regenerative responses.


Subject(s)
Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Proliferation/physiology , Cells, Cultured , Genes, fos , Mice
2.
Nat Commun ; 11(1): 586, 2020 Jan 29.
Article in English | MEDLINE | ID: mdl-31996681

ABSTRACT

The endothelial to haematopoietic transition (EHT) is the process whereby haemogenic endothelium differentiates into haematopoietic stem and progenitor cells (HSPCs). The intermediary steps of this process are unclear, in particular the identity of endothelial cells that give rise to HSPCs is unknown. Using single-cell transcriptome analysis and antibody screening, we identify CD44 as a marker of EHT enabling us to isolate robustly the different stages of EHT in the aorta-gonad-mesonephros (AGM) region. This allows us to provide a detailed phenotypical and transcriptional profile of CD44-positive arterial endothelial cells from which HSPCs emerge. They are characterized with high expression of genes related to Notch signalling, TGFbeta/BMP antagonists, a downregulation of genes related to glycolysis and the TCA cycle, and a lower rate of cell cycle. Moreover, we demonstrate that by inhibiting the interaction between CD44 and its ligand hyaluronan, we can block EHT, identifying an additional regulator of HSPC development.


Subject(s)
Biomarkers , Endothelium/metabolism , Hematopoietic Stem Cells/metabolism , Hyaluronan Receptors/metabolism , Transcriptome , Animals , Aorta , Arteries , Cell Cycle , Citric Acid Cycle/genetics , Computational Biology , Core Binding Factor Alpha 2 Subunit/genetics , Down-Regulation , Glycolysis/genetics , Gonads , Hematopoiesis/physiology , Hyaluronan Receptors/blood , Hyaluronan Receptors/genetics , Hyaluronic Acid , Mesonephros , Mice , Mice, Inbred C57BL , Mice, Knockout , Transforming Growth Factor beta/metabolism
3.
JCI Insight ; 52019 04 30.
Article in English | MEDLINE | ID: mdl-31039138

ABSTRACT

Monosomy 7 or deletion of 7q (del(7q)) are common clonal cytogenetic abnormalities associated with high grade myelodysplastic syndrome (MDS) arising in inherited and acquired bone marrow failure. Current non-transplant approaches to treat marrow failure may be complicated by stimulation of clonal outgrowth. To study the biological consequences of del(7q) within the context of a failing marrow, we generated induced pluripotent stem cells (iPSCs) derived from patients with Shwachman Diamond Syndrome (SDS), a bone marrow failure disorder with MDS predisposition, and genomically engineered a 7q deletion. The TGFß pathway was the top differentially regulated pathway in transcriptomic analysis of SDS versus SDSdel(7q) iPSCs. SMAD2 phosphorylation was increased in SDS relative to wild type cells consistent with hyperactivation of the TGFbeta pathway in SDS. Phospho-SMAD2 levels were reduced following 7q deletion in SDS cells and increased upon restoration of 7q diploidy. Inhibition of the TGFbeta pathway rescued hematopoiesis in SDS-iPSCs and in bone marrow hematopoietic cells from SDS patients while it had no impact on the SDSdel(7q) cells. These results identified a potential targetable vulnerability to improve hematopoiesis in an MDS-predisposition syndrome, and highlight the importance of the germline context of somatic alterations to inform precision medicine approaches to therapy.


Subject(s)
Bone Marrow/pathology , Myelodysplastic Syndromes/prevention & control , Precision Medicine/methods , Shwachman-Diamond Syndrome/therapy , Bone Marrow/drug effects , Cell Engineering , Chromosome Deletion , Chromosomes, Human, Pair 7/genetics , HEK293 Cells , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/pathology , Karyotyping , Myelodysplastic Syndromes/genetics , Phosphorylation/genetics , RNA-Seq , Shwachman-Diamond Syndrome/diagnosis , Shwachman-Diamond Syndrome/genetics , Shwachman-Diamond Syndrome/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism
4.
J Clin Invest ; 129(9): 3821-3826, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31211692

ABSTRACT

Shwachman-Diamond Syndrome (SDS) is a rare and clinically-heterogeneous bone marrow (BM) failure syndrome caused by mutations in the Shwachman-Bodian-Diamond Syndrome (SBDS) gene. Although SDS was described over 50 years ago, the molecular pathogenesis is poorly understood due, in part, to the rarity and heterogeneity of the affected hematopoietic progenitors. To address this, we used single cell RNA sequencing to profile scant hematopoietic stem and progenitor cells from SDS patients. We generated a single cell map of early lineage commitment and found that SDS hematopoiesis was left-shifted with selective loss of granulocyte-monocyte progenitors. Transcriptional targets of transforming growth factor-beta (TGFß) were dysregulated in SDS hematopoietic stem cells and multipotent progenitors, but not in lineage-committed progenitors. TGFß inhibitors (AVID200 and SD208) increased hematopoietic colony formation of SDS patient BM. Finally, TGFß3 and other TGFß pathway members were elevated in SDS patient blood plasma. These data establish the TGFß pathway as a novel candidate biomarker and therapeutic target in SDS and translate insights from single cell biology into a potential therapy.


Subject(s)
Bone Marrow/physiopathology , Hematopoietic Stem Cells/pathology , Shwachman-Diamond Syndrome/physiopathology , Transforming Growth Factor beta1/metabolism , Adolescent , Adult , Antigens, CD34/metabolism , Cell Differentiation , Cell Lineage , Child , Granulocytes/cytology , Hematopoiesis , Humans , Inflammation , Monocytes/cytology , Mutation , Phosphorylation , Sequence Analysis, RNA , Signal Transduction , Transforming Growth Factor beta/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL