Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
Blood ; 138(17): 1603-1614, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34115825

ABSTRACT

EZH2, the enzymatic component of PRC2, has been identified as a key factor in hematopoiesis. EZH2 loss-of-function mutations have been found in myeloproliferative neoplasms, particularly in myelofibrosis, but the precise function of EZH2 in megakaryopoiesis is not fully delineated. Here, we show that EZH2 inhibition by small molecules and short hairpin RNA induces megakaryocyte (MK) commitment by accelerating lineage marker acquisition without change in proliferation. Later in differentiation, EZH2 inhibition blocks proliferation and polyploidization and decreases proplatelet formation. EZH2 inhibitors similarly reduce MK polyploidization and proplatelet formation in vitro and platelet levels in vivo in a JAK2V617F background. In transcriptome profiling, the defect in proplatelet formation was associated with an aberrant actin cytoskeleton regulation pathway, whereas polyploidization was associated with an inhibition of expression of genes involved in DNA replication and repair and an upregulation of cyclin-dependent kinase inhibitors, particularly CDKN1A and CDKN2D. The knockdown of CDKN1A and to a lesser extent CDKN2D could partially rescue the percentage of polyploid MKs. Moreover, H3K27me3 and EZH2 chromatin immunoprecipitation assays revealed that CDKN1A is a direct EZH2 target and CDKN2D expression is not directly regulated by EZH2, suggesting that EZH2 controls MK polyploidization directly through CDKN1A and indirectly through CDKN2D.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Megakaryocytes/cytology , Thrombopoiesis , Animals , Blood Platelets/cytology , Blood Platelets/metabolism , Cells, Cultured , Enhancer of Zeste Homolog 2 Protein/genetics , Humans , Megakaryocytes/metabolism , Mice , RNA Interference , Transcriptome
3.
Stem Cell Reports ; 13(2): 274-290, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31327733

ABSTRACT

Bone marrow (BM) niche cells help to keep adult hematopoietic stem cells (HSCs) in a quiescent state via secreted factors and induction of cell-cycle inhibitors. Here, we demonstrate that the adapter protein CABLES1 is a key regulator of long-term hematopoietic homeostasis during stress and aging. Young mice lacking Cables1 displayed hyperproliferation of hematopoietic progenitor cells. This defect was cell intrinsic, since it was reproduced in BM transplantation assays using wild-type animals as recipients. Overexpression and short hairpin RNA-mediated depletion of CABLES1 protein resulted in p21Cip/waf up- and downregulation, respectively. Aged mice lacking Cables1 displayed abnormalities in peripheral blood cell counts accompanied by a significant reduction in HSC compartment, concomitant with an increased mobilization of progenitor cells. In addition, Cables1-/- mice displayed increased sensitivity to the chemotherapeutic agent 5-fluorouracil due to an abnormal microenvironment. Altogether, our findings uncover a key role for CABLES1 in HSC homeostasis and stress hematopoiesis.


Subject(s)
Cyclins/genetics , Hematopoietic Stem Cells/cytology , Aging , Animals , Antigens, CD34/metabolism , Bone Marrow Cells/cytology , Cell Cycle Checkpoints/drug effects , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclins/antagonists & inhibitors , Cyclins/metabolism , Fluorouracil/pharmacology , Hematopoiesis/drug effects , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA Interference , RNA, Small Interfering/metabolism
4.
Cell Stem Cell ; 25(3): 407-418.e6, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31303548

ABSTRACT

Hematopoietic stem cells (HSCs) residing in the bone marrow (BM) accumulate during aging but are functionally impaired. However, the role of HSC-intrinsic and -extrinsic aging mechanisms remains debated. Megakaryocytes promote quiescence of neighboring HSCs. Nonetheless, whether megakaryocyte-HSC interactions change during pathological/natural aging is unclear. Premature aging in Hutchinson-Gilford progeria syndrome recapitulates physiological aging features, but whether these arise from altered stem or niche cells is unknown. Here, we show that the BM microenvironment promotes myelopoiesis in premature/physiological aging. During physiological aging, HSC-supporting niches decrease near bone but expand further from bone. Increased BM noradrenergic innervation promotes ß2-adrenergic-receptor(AR)-interleukin-6-dependent megakaryopoiesis. Reduced ß3-AR-Nos1 activity correlates with decreased endosteal niches and megakaryocyte apposition to sinusoids. However, chronic treatment of progeroid mice with ß3-AR agonist decreases premature myeloid and HSC expansion and restores the proximal association of HSCs to megakaryocytes. Therefore, normal/premature aging of BM niches promotes myeloid expansion and can be improved by targeting the microenvironment.


Subject(s)
Aging, Premature/pathology , Aging/physiology , Bone Marrow/physiology , Hematopoietic Stem Cells/physiology , Megakaryocytes/physiology , Myeloid Cells/physiology , Progeria/pathology , Adrenergic Agonists/administration & dosage , Aging/metabolism , Aging, Premature/metabolism , Animals , Cell Differentiation , Cell Encapsulation , Cell Proliferation , Disease Models, Animal , Humans , Interleukin-6/metabolism , Mice , Nitric Oxide Synthase Type I/metabolism , Progeria/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction , Stem Cell Niche
5.
Blood Adv ; 3(4): 633-643, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30804017

ABSTRACT

Antibody-drug conjugates (ADCs) are a new class of therapeutics that use antibodies to deliver potent cytotoxic drugs selectively to cancer cells. CD203c, an ecto-nucleotide pyrophosphatase-phosphodiesterase 3, is overexpressed on neoplastic mast cells (MCs) in systemic mastocytosis (SM), thus representing a promising target for antibody-mediated therapy. In this study, we have found that human neoplastic MC lines (ROSAKIT D816V and ROSAKIT D816V-Gluc), which express high levels of CD203c, are highly and specifically sensitive to the antiproliferative effects of an ADC against CD203c (AGS-16C3F). In these cell lines, AGS-16C3F induced cell apoptosis at very low concentrations. To characterize the effects of AGS-16C3F on leukemia progression in vivo, ROSAKIT D816V-Gluc NOD-SCID γ mouse models of advanced SM (AdvSM) were treated with AGS-16C3F or an ADC control for 2 weeks. Whereas AGS-16C3F had no apparent toxicity in xenotransplanted mice, in vivo neoplastic MC burden significantly decreased in both hematopoietic and nonhematopoietic organs. Furthermore, animals treated with AGS-16C3F had prolonged survival compared with the animals treated with control ADC, and AGS-16C3F efficiently prevented disease relapse. In conclusion, these preclinical studies identified CD203c as a novel therapeutic target on neoplastic MCs, and AGS-16C3F as a promising ADC for the treatment of patients with AdvSM.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Immunoconjugates/therapeutic use , Mastocytosis, Systemic/drug therapy , Animals , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Humans , Mastocytosis, Systemic/pathology , Mice , Mice, Inbred NOD , Mice, SCID
6.
Oncotarget ; 8(33): 54082-54095, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28903325

ABSTRACT

JAK2 activation is the driver mechanism in BCR-ABL-negative myeloproliferative neoplasms (MPN). These diseases are characterized by an abnormal retention of hematopoietic stem cells within the bone marrow microenvironment and their increased trafficking to extramedullary sites. The CXCL12/CXCR4 axis plays a central role in hematopoietic stem cell/ progenitor trafficking and retention in hematopoietic sites. The present study explores the crosstalk between JAK2 and CXCL12/CXCR4 signaling pathways in MPN. We show that JAK2, activated by either MPL-W515L expression or cytokine stimulation, cooperates with CXCL12/CXCR4 signaling to increase the chemotactic response of human cell lines and primary CD34+ cells through an increased phosphatidylinositol-3-kinase (PI3K) signaling. Accordingly, primary myelofibrosis (MF) patient cells demonstrate an increased CXCL12-induced chemotaxis when compared to controls. JAK2 inhibition by knock down or chemical inhibitors decreases this effect in MPL-W515L expressing cell lines and reduces the CXCL12/CXCR4 signaling in some patient primary cells. Taken together, these data indicate that CXCL12/CXCR4 pathway is overactivated in MF patients by oncogenic JAK2 that maintains high PI3K signaling over the threshold required for CXCR4 activation. These results suggest that inhibition of this crosstalk may contribute to the therapeutic effects of JAK2 inhibitors.

7.
J Exp Med ; 214(7): 2023-2040, 2017 Jul 03.
Article in English | MEDLINE | ID: mdl-28550161

ABSTRACT

The CXCL12/CXCR4 signaling exerts a dominant role in promoting hematopoietic stem and progenitor cell (HSPC) retention and quiescence in bone marrow. Gain-of-function CXCR4 mutations that affect homologous desensitization of the receptor have been reported in the WHIM Syndrome (WS), a rare immunodeficiency characterized by lymphopenia. The mechanisms underpinning this remain obscure. Using a mouse model with a naturally occurring WS-linked gain-of-function Cxcr4 mutation, we explored the possibility that the lymphopenia in WS arises from defects at the HSPC level. We reported that Cxcr4 desensitization is required for quiescence/cycling balance of murine short-term hematopoietic stem cells and their differentiation into multipotent and downstream lymphoid-biased progenitors. Alteration in Cxcr4 desensitization resulted in decrease of circulating HSPCs in five patients with WS. This was also evidenced in WS mice and mirrored by accumulation of HSPCs in the spleen, where we observed enhanced extramedullary hematopoiesis. Therefore, efficient Cxcr4 desensitization is critical for lymphoid differentiation of HSPCs, and its impairment is a key mechanism underpinning the lymphopenia observed in mice and likely in WS patients.


Subject(s)
Cell Differentiation/genetics , Hematopoietic Stem Cells/metabolism , Lymphocytes/metabolism , Receptors, CXCR4/genetics , Adult , Animals , Bone Marrow Cells/metabolism , Bone Marrow Transplantation/methods , Cell Survival/genetics , Child , Flow Cytometry , Gene Expression , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism , Lymphocyte Count , Mice, Transgenic , Mutation , Primary Immunodeficiency Diseases , Receptors, CXCR4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Spleen/cytology , Spleen/metabolism , Warts/genetics , Warts/metabolism
8.
Sci Rep ; 6: 37827, 2016 11 25.
Article in English | MEDLINE | ID: mdl-27886253

ABSTRACT

Hematopoietic stem cells (HSCs) undergo self-renewal to maintain hematopoietic homeostasis for lifetime, which is regulated by the bone marrow (BM) microenvironment. The chemokine receptor CXCR4 and its ligand CXCL12 are critical factors supporting quiescence and BM retention of HSCs. Here, we report an unknown function of CXCR4/CXCL12 axis in the protection of HSCs against oxidative stress. Disruption of CXCR4 receptor in mice leads to increased endogenous production of reactive oxygen species (ROS), resulting in p38 MAPK activation, increased DNA double-strand breaks and apoptosis leading to marked reduction in HSC repopulating potential. Increased ROS levels are directly responsible for exhaustion of the HSC pool and are not linked to loss of quiescence of CXCR4-deficient HSCs. Furthermore, we report that CXCL12 has a direct rescue effect on oxidative stress-induced HSC damage at the mitochondrial level. These data highlight the importance of CXCR4/CXCL12 axis in the regulation of lifespan of HSCs by limiting ROS generation and genotoxic stress.


Subject(s)
Chemokine CXCL12/genetics , Hematopoietic Stem Cells/cytology , Hepatocytes/cytology , Reactive Oxygen Species/metabolism , Receptors, CXCR4/genetics , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chemokine CXCL12/metabolism , DNA Breaks, Double-Stranded , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Hepatocytes/metabolism , Mice , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Stress , Phosphorylation , Receptors, CXCR4/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Stem Cells ; 25(1): 211-9, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17023512

ABSTRACT

As an intracellular second messenger, nitric oxide (NO) is increasingly implicated in the control of transcriptional machinery and gene expression. Here, we show that cell surface expression of CXCR4 on CD34(+) cells was increased in a dose- and time-dependent manner in response to NO donors. Augmented surface expression was correlated with an increase in CXCR4 mRNA level. A specific NO scavenger prevented the elevation in CXCR4 mRNA caused by NO donors, suggesting a direct signaling action mediated by NO on CXCR4 transcription. NO treatment had no significant effect on CXCR4 mRNA stability. However, induction of CXCR4 mRNA by NO was still observed in conditions in which initiation of translation was inhibited, suggesting that the NO effect must be mediated by a pre-existing protein. CXCR4 mRNA induction did not involve cGMP (guanosine 3', 5'-cyclic monophosphate) generation but was most likely mediated via oxidation of intracellular protein thiols. Finally, CD34(+) cells pretreated with NO donors exhibited an increased chemotactic response. This study demonstrates that the NO pathway can modulate CXCR4 expression in human CD34(+) cells and suggests that NO may play a critical role in the trafficking of hematopoietic progenitors.


Subject(s)
Antigens, CD34/analysis , Gene Expression Regulation/physiology , Hematopoietic Stem Cells/physiology , Nitric Oxide Donors/pharmacology , Nitric Oxide/pharmacology , Receptors, CXCR4/genetics , Cell Culture Techniques , Cell Survival/drug effects , DNA Primers , Fetal Blood/cytology , Flow Cytometry , Gene Expression Regulation/drug effects , Hematopoietic Stem Cells/drug effects , Humans , Infant, Newborn , Kinetics , Nitroso Compounds/pharmacology , Polymerase Chain Reaction , RNA, Messenger/genetics , Transcription, Genetic/drug effects
10.
Blood ; 107(6): 2243-51, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16291599

ABSTRACT

The physiologic role of CXCR4 on hematopoietic stem/progenitor cells (HSPCs) is not fully understood. Here, we show that radioprotection of lethally irradiated mice by embryonic day 14.5 (E14.5) CXCR4-/- fetal liver (FL) cells was markedly impaired when compared with CXCR4+/+ counterparts, but this defect was rescued when hosts were engrafted with high cell numbers. This quantitative defect contrasted with a similar content in hematopoietic colony-forming cells (CFCs), splenic colony-forming units (CFUs-S), and Lin- Sca-1+ c-kit+ cells in E14.5 CXCR4-/- and CXCR4+/+ livers. In addition, the homing of HSPCs in the bone marrow was not altered as detected with a CFSE-staining assay. In contrast, a 30-fold increase in CFCs was seen in the circulation of mice stably reconstituted with CXCR4-/- FL cells and this increment was already observed before hematopoiesis had reached a steady-state level. Together, the data strongly suggest that impaired retention may, at least in short-term hematopoietic reconstitution, lead to a diminution in the number of available progenitors required for radioprotection.


Subject(s)
Bone Marrow/physiology , Hematopoietic Stem Cells/physiology , Whole-Body Irradiation , Animals , Cell Movement , Embryo, Mammalian , Hematopoiesis , Liver/cytology , Mice , Mice, Knockout , Mice, Transgenic , Receptors, CXCR4 , Spleen/cytology
11.
Proc Natl Acad Sci U S A ; 100(23): 13710-5, 2003 Nov 11.
Article in English | MEDLINE | ID: mdl-14595018

ABSTRACT

To gain a molecular understanding of kidney functions, we established a high-resolution map of gene expression patterns in the human kidney. The glomerulus and seven different nephron segments were isolated by microdissection from fresh tissue specimens, and their transcriptome was characterized by using the serial analysis of gene expression (SAGE) method. More than 400,000 mRNA SAGE tags were sequenced, making it possible to detect in each structure transcripts present at 18 copies per cell with a 95% confidence level. Expression of genes responsible for nephron transport and permeability properties was evidenced through transcripts for 119 solute carriers, 84 channels, 43 ion-transport ATPases, and 12 claudins. Searching for differences between the transcriptomes, we found 998 transcripts greatly varying in abundance from one nephron portion to another. Clustering analysis of these transcripts evidenced different extents of similarity between the nephron portions. Approximately 75% of the differentially distributed transcripts corresponded to cDNAs of known or unknown function that are accurately mapped in the human genome. This systematic large-scale analysis of individual structures of a complex human tissue reveals sets of genes underlying the function of well-defined nephron portions. It also provides quantitative expression data for a variety of genes mutated in hereditary diseases and helps in sorting candidate genes for renal diseases that affect specific portions of the human nephron.


Subject(s)
Gene Expression , Kidney/metabolism , Oligonucleotide Array Sequence Analysis , Cluster Analysis , DNA, Complementary/metabolism , Gene Library , Humans , Models, Anatomic , Molecular Sequence Data , Nephrons/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Software
12.
J Am Soc Nephrol ; 12(3): 598-601, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181809

ABSTRACT

Micropuncture studies of the distal nephron and measurements of Na,K-ATPase activity in microdissected collecting tubules have suggested that renal retention of sodium in puromycin aminonucleoside (PAN) nephrotic rats originates in the collecting duct. The present study demonstrated this hypothesis by in vitro microperfusion and showed that amiloride was able to restore sodium balance. Indeed, isolated perfused cortical collecting ducts from PAN-treated rats exhibited an abnormally high transepithelial sodium reabsorption that was abolished by amiloride, and in vivo administration of amiloride fully prevented decreased urinary sodium excretion and positive sodium balance in nephrotic rats. As expected from the aldosterone independence of Na(+) retention in PAN nephrotic rats, blockade of aldosterone receptor by potassium canrenoate did not alter urinary Na(+) excretion, Na(+) balance, or ascites formation in PAN nephrotic rats.


Subject(s)
Amiloride/therapeutic use , Kidney Tubules, Collecting/metabolism , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/metabolism , Sodium/metabolism , Animals , Canrenoic Acid/therapeutic use , In Vitro Techniques , Kidney Tubules, Collecting/drug effects , Male , Mineralocorticoid Receptor Antagonists/therapeutic use , Natriuresis/drug effects , Nephrotic Syndrome/chemically induced , Perfusion , Puromycin Aminonucleoside/toxicity , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers
SELECTION OF CITATIONS
SEARCH DETAIL
...