Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 16 de 16
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
JCI Insight ; 9(4)2024 Jan 11.
Article de Anglais | MEDLINE | ID: mdl-38385749

RÉSUMÉ

RNA-binding proteins (RBPs) interact with RNA and ubiquitously regulate RNA transcripts during their life cycle, playing a fundamental role in the progression of angiogenesis-related diseases. In the skeletal system, endothelium-dependent angiogenesis is indispensable for bone formation. However, the role of RBPs in endothelium-dependent bone formation is unclear. Here, we show that RBP-Y-box-binding protein 1 (YBX1) was strongly reduced in the bone vasculature of ovariectomy (OVX) mice. Endothelial cell-specific deletion of Ybx1 impaired CD31-high, endomucin-high (CD31hiEMCNhi) endothelium morphology and resulted in low bone mass whereas Ybx1 overexpression promoted angiogenesis-dependent osteogenesis and ameliorated bone loss. Mechanistically, YBX1 deletion disrupted CD31, EMCN, and bone morphogenetic protein 4 (BMP4) stability in an m5C-dependent manner and blocked endothelium-derived BMP4 release, thereby inhibiting osteogenic differentiation of bone mesenchymal stromal cells. Administration of recombinant BMP4 protein restored impaired bone formation in Ybx1 deletion mice. Tail vein injection of CD31-modified polyethylene glycol-poly (lactic-co-glycolic acid) carrying sciadopitysin, a natural YBX1 agonist, pharmacologically partially reversed CD31hiEMCNhi vessels' decline and improved bone mass in both OVX and aging animals. These findings demonstrated the role of RBP-YBX1 in angiogenesis-dependent bone formation and provided a therapeutic approach for ameliorating osteoporosis.


Sujet(s)
Ostéogenèse , Ostéoporose , Facteurs de transcription , Animaux , Femelle , Souris , Os et tissu osseux/métabolisme , Endothélium/métabolisme , Ostéogenèse/physiologie , Ostéoporose/génétique , ARN , Facteurs de transcription/génétique
2.
J Clin Invest ; 133(15)2023 08 01.
Article de Anglais | MEDLINE | ID: mdl-37526082

RÉSUMÉ

Clonal hematopoiesis plays a critical role in the initiation and development of hematologic malignancies. In patients with del(5q) myelodysplastic syndrome (MDS), the transcription factor FOXM1 is frequently downregulated in CD34+ cells. In this study, we demonstrated that Foxm1 haploinsufficiency disturbed normal hematopoiesis and conferred a competitive repopulation advantage for a short period. However, it impaired the long-term self-renewal capacity of hematopoietic stem cells, recapitulating the phenotypes of abnormal hematopoietic stem cells observed in patients with MDS. Moreover, heterozygous inactivation of Foxm1 led to an increase in DNA damage in hematopoietic stem/progenitor cells (HSPCs). Foxm1 haploinsufficiency induced hematopoietic dysplasia in a mouse model with LPS-induced chronic inflammation and accelerated AML-ETO9a-mediated leukemogenesis. We have also identified Parp1, an important enzyme that responds to various types of DNA damage, as a target of Foxm1. Foxm1 haploinsufficiency decreased the ability of HSPCs to efficiently repair DNA damage by downregulating Parp1 expression. Our findings suggest that the downregulation of the Foxm1-Parp1 molecular axis may promote clonal hematopoiesis and reduce genome stability, contributing to del(5q) MDS pathogenesis.


Sujet(s)
Hématopoïèse clonale , Protéine M1 à motif en tête de fourche , Tumeurs hématologiques , Animaux , Souris , Protéine M1 à motif en tête de fourche/génétique , Tumeurs hématologiques/génétique , Tumeurs hématologiques/anatomopathologie , Cellules souches hématopoïétiques , Altération de l'ADN , Poly (ADP-Ribose) polymerase-1/métabolisme , Souris de lignée C57BL
3.
JCI Insight ; 8(1)2023 01 10.
Article de Anglais | MEDLINE | ID: mdl-36378535

RÉSUMÉ

The central physiological role of the bone marrow renders bone marrow stromal cells (BMSCs) particularly sensitive to aging. With bone aging, BMSCs acquire a differentiation potential bias in favor of adipogenesis over osteogenesis, and the underlying molecular mechanisms remain unclear. Herein, we investigated the factors underlying age-related changes in the bone marrow and their roles in BMSCs' differentiation. Antibody array revealed that CC chemokine ligand 3 (CCL3) accumulation occurred in the serum of naturally aged mice along with bone aging phenotypes, including bone loss, bone marrow adiposity, and imbalanced BMSC differentiation. In vivo Ccl3 deletion could rescue these phenotypes in aged mice. CCL3 improved the adipogenic differentiation potential of BMSCs, with a positive feedback loop between CCL3 and C/EBPα. CCL3 activated C/EBPα expression via STAT3, while C/EBPα activated CCL3 expression through direct promoter binding, facilitated by DNA hypomethylation. Moreover, CCL3 inhibited BMSCs' osteogenic differentiation potential by blocking ß-catenin activity mediated by ERK-activated Dickkopf-related protein 1 upregulation. Blocking CCL3 in vivo via neutralizing antibodies ameliorated trabecular bone loss and bone marrow adiposity in aged mice. This study provides insights regarding age-related bone loss and bone marrow adiposity pathogenesis and lays a foundation for the identification of new targets for senile osteoporosis treatment.


Sujet(s)
Ostéogenèse , Ostéoporose , Souris , Animaux , Ostéogenèse/physiologie , Adiposité , Moelle osseuse/anatomopathologie , Ligands , Différenciation cellulaire , Ostéoporose/métabolisme , Obésité/complications , Chimiokine CCL3/génétique
4.
JCI Insight ; 7(24)2022 12 22.
Article de Anglais | MEDLINE | ID: mdl-36546480

RÉSUMÉ

Primary atopic disorders are a group of inborn errors of immunity that skew the immune system toward severe allergic disease. Defining the biology underlying these extreme monogenic phenotypes reveals shared mechanisms underlying common polygenic allergic disease and identifies potential drug targets. Germline gain-of-function (GOF) variants in JAK1 are a cause of severe atopy and eosinophilia. Modeling the JAK1GOF (p.A634D) variant in both zebrafish and human induced pluripotent stem cells (iPSCs) revealed enhanced myelopoiesis. RNA-Seq of JAK1GOF human whole blood, iPSCs, and transgenic zebrafish revealed a shared core set of dysregulated genes involved in IL-4, IL-13, and IFN signaling. Immunophenotypic and transcriptomic analysis of patients carrying a JAK1GOF variant revealed marked Th cell skewing. Moreover, long-term ruxolitinib treatment of 2 children carrying the JAK1GOF (p.A634D) variant remarkably improved their growth, eosinophilia, and clinical features of allergic inflammation. This work highlights the role of JAK1 signaling in atopic immune dysregulation and the clinical impact of JAK1/2 inhibition in treating eosinophilic and allergic disease.


Sujet(s)
Éosinophilie , Hypersensibilité immédiate , Hypersensibilité , Cellules souches pluripotentes induites , Enfant , Animaux , Humains , Mutation gain de fonction , Danio zébré , Hypersensibilité/génétique , Inflammation/génétique , Éosinophilie/génétique , Janus kinase 1/génétique
5.
J Clin Invest ; 132(19)2022 10 03.
Article de Anglais | MEDLINE | ID: mdl-35925681

RÉSUMÉ

Infantile (fetal and neonatal) megakaryocytes (Mks) have a distinct phenotype consisting of hyperproliferation, limited morphogenesis, and low platelet production capacity. These properties contribute to clinical problems that include thrombocytopenia in neonates, delayed platelet engraftment in recipients of cord blood stem cell transplants, and inefficient ex vivo platelet production from pluripotent stem cell-derived Mks. The infantile phenotype results from deficiency of the actin-regulated coactivator, MKL1, which programs cytoskeletal changes driving morphogenesis. As a strategy to complement this molecular defect, we screened pathways with the potential to affect MKL1 function and found that DYRK1A inhibition dramatically enhanced Mk morphogenesis in vitro and in vivo. Dyrk1 inhibitors rescued enlargement, polyploidization, and thrombopoiesis in human neonatal Mks. Mks derived from induced pluripotent stem cells responded in a similar manner. Progenitors undergoing Dyrk1 inhibition demonstrated filamentous actin assembly, MKL1 nuclear translocation, and modulation of MKL1 target genes. Loss-of-function studies confirmed MKL1 involvement in this morphogenetic pathway. Expression of Ablim2, a stabilizer of filamentous actin, increased with Dyrk1 inhibition, and Ablim2 knockdown abrogated the actin, MKL1, and morphogenetic responses to Dyrk1 inhibition. These results delineate a pharmacologically tractable morphogenetic pathway whose manipulation may alleviate clinical problems associated with the limited thrombopoietic capacity of infantile Mks.


Sujet(s)
Mégacaryocytes , Thrombopénie , Actines/métabolisme , Plaquettes/métabolisme , Humains , Nouveau-né , Mégacaryocytes/métabolisme , Phénotype , Protein-Serine-Threonine Kinases , Protein-tyrosine kinases , Thrombopénie/génétique , Thrombopoïèse/génétique ,
6.
JCI Insight ; 6(24)2021 12 22.
Article de Anglais | MEDLINE | ID: mdl-34793338

RÉSUMÉ

The clinical utility of histone/protein deacetylase (HDAC) inhibitors in combinatorial regimens with proteasome inhibitors for patients with relapsed and refractory multiple myeloma (MM) is often limited by excessive toxicity due to HDAC inhibitor promiscuity with multiple HDACs. Therefore, more selective inhibition minimizing off-target toxicity may increase the clinical effectiveness of HDAC inhibitors. We demonstrated that plasma cell development and survival are dependent upon HDAC11, suggesting this enzyme is a promising therapeutic target in MM. Mice lacking HDAC11 exhibited markedly decreased plasma cell numbers. Accordingly, in vitro plasma cell differentiation was arrested in B cells lacking functional HDAC11. Mechanistically, we showed that HDAC11 is involved in the deacetylation of IRF4 at lysine103. Further, targeting HDAC11 led to IRF4 hyperacetylation, resulting in impaired IRF4 nuclear localization and target promoter binding. Importantly, transient HDAC11 knockdown or treatment with elevenostat, an HDAC11-selective inhibitor, induced cell death in MM cell lines. Elevenostat produced similar anti-MM activity in vivo, improving survival among mice inoculated with 5TGM1 MM cells. Elevenostat demonstrated nanomolar ex vivo activity in 34 MM patient specimens and synergistic activity when combined with bortezomib. Collectively, our data indicated that HDAC11 regulates an essential pathway in plasma cell biology establishing its potential as an emerging theraputic vulnerability in MM.


Sujet(s)
Inhibiteurs de désacétylase d'histone/usage thérapeutique , Histone/métabolisme , Myélome multiple/traitement médicamenteux , Plasmocytes/métabolisme , Animaux , Inhibiteurs de désacétylase d'histone/pharmacologie , Humains , Souris , Myélome multiple/physiopathologie
7.
JCI Insight ; 6(19)2021 10 08.
Article de Anglais | MEDLINE | ID: mdl-34622798

RÉSUMÉ

Hypomorphic RAG1 or RAG2 mutations cause primary immunodeficiencies and can lead to autoimmunity, but the underlying mechanisms are elusive. We report here a patient carrying a c.116+2T>G homozygous splice site mutation in the first intron of RAG1, which led to aberrant splicing and greatly reduced RAG1 protein expression. B cell development was blocked at both the pro-B to pre-B transition and the pre-B to immature B cell differentiation step. The patient B cells had reduced B cell receptor repertoire diversity and decreased complementarity determining region 3 lengths. Despite B cell lymphopenia, the patient had abundant plasma cells in the BM and produced large quantities of IgM and IgG Abs, including autoantibodies. The proportion of naive B cells was reduced while the frequency of IgD-CD27- double-negative (DN) B cells, which quickly differentiated into Ab-secreting plasma cells upon stimulation, was greatly increased. Immune phenotype analysis of 52 patients with primary immunodeficiency revealed a strong association of the increased proportion of DN B and memory B cells with decreased number and proportion of naive B cells. These results suggest that the lymphopenic environment triggered naive B cell differentiation into DN B and memory B cells, leading to increased Ab production.


Sujet(s)
Autoanticorps/immunologie , Maladies auto-immunes/génétique , Lymphocytes B/immunologie , Granulome/génétique , Protéines à homéodomaine/génétique , Déficits immunitaires/génétique , Lymphopoïèse/génétique , Récepteurs pour l'antigène des lymphocytes B/immunologie , Maladies auto-immunes/immunologie , Maladies auto-immunes/thérapie , Enfant , Transplantation de cellules souches de sang du cordon , Issue fatale , Granulome/immunologie , Granulome/thérapie , Protéines à homéodomaine/métabolisme , Homozygote , Humains , Immunoglobuline G/immunologie , Immunoglobuline M/immunologie , Déficits immunitaires/immunologie , Déficits immunitaires/thérapie , Mémoire immunologique/immunologie , Lymphopénie/génétique , Lymphopénie/immunologie , Lymphopoïèse/immunologie , Mâle , Plasmocytes/immunologie , Sites d'épissage d'ARN/génétique , Recombinaison V(D)J/génétique
8.
JCI Insight ; 6(16)2021 08 23.
Article de Anglais | MEDLINE | ID: mdl-34255742

RÉSUMÉ

Neutrophils are produced in the BM in a process called granulopoiesis, in which progenitor cells sequentially develop into mature neutrophils. During the developmental process, which is finely regulated by distinct transcription factors, neutrophils acquire the ability to exit the BM, properly distribute throughout the body, and migrate to infection sites. Previous studies have demonstrated that CD40 ligand (CD40L) influences hematopoiesis and granulopoiesis. Here, we investigate the effect of CD40L on neutrophil development and trafficking by performing functional and transcriptome analyses. We found that CD40L signaling plays an essential role in the early stages of neutrophil generation and development in the BM. Moreover, CD40L modulates transcriptional signatures, indicating that this molecule enables neutrophils to traffic throughout the body and to migrate in response to inflammatory signals. Thus, our study provides insights into the complex relationships between CD40L signaling and granulopoiesis, and it suggests a potentially novel and nonredundant role of CD40L signaling in neutrophil development and function.


Sujet(s)
Moelle osseuse/croissance et développement , Antigènes CD40/métabolisme , Ligand de CD40/métabolisme , Hématopoïèse/génétique , Granulocytes neutrophiles/physiologie , Animaux , Ligand de CD40/génétique , Différenciation cellulaire/génétique , Mouvement cellulaire/génétique , Cellules cultivées , Femelle , Régulation de l'expression des gènes au cours du développement , Souris , Souris knockout , Modèles animaux , RNA-Seq , Transduction du signal/génétique
9.
J Clin Invest ; 131(2)2021 01 19.
Article de Anglais | MEDLINE | ID: mdl-33206630

RÉSUMÉ

Bone is maintained by coupled activities of bone-forming osteoblasts/osteocytes and bone-resorbing osteoclasts. Alterations in this relationship can lead to pathologic bone loss such as osteoporosis. It is well known that osteogenic cells support osteoclastogenesis via production of RANKL. Interestingly, our recently identified bone marrow mesenchymal cell population-marrow adipogenic lineage precursors (MALPs) that form a multidimensional cell network in bone-was computationally demonstrated to be the most interactive with monocyte-macrophage lineage cells through high and specific expression of several osteoclast regulatory factors, including RANKL. Using an adipocyte-specific Adipoq-Cre to label MALPs, we demonstrated that mice with RANKL deficiency in MALPs have a drastic increase in trabecular bone mass in long bones and vertebrae starting from 1 month of age, while their cortical bone appears normal. This phenotype was accompanied by diminished osteoclast number and attenuated bone formation at the trabecular bone surface. Reduced RANKL signaling in calvarial MALPs abolished osteolytic lesions after LPS injections. Furthermore, in ovariectomized mice, elevated bone resorption was partially attenuated by RANKL deficiency in MALPs. In summary, our studies identified MALPs as a critical player in controlling bone remodeling during normal bone metabolism and pathological bone loss in a RANKL-dependent fashion.


Sujet(s)
Moelle osseuse , Remodelage osseux , Résorption osseuse , Ostéoclastes , Adipocytes/métabolisme , Adipocytes/anatomopathologie , Adiponectine/génétique , Adiponectine/métabolisme , Animaux , Moelle osseuse/métabolisme , Moelle osseuse/anatomopathologie , Résorption osseuse/génétique , Résorption osseuse/métabolisme , Résorption osseuse/anatomopathologie , Souris , Souris transgéniques , Ostéoclastes/métabolisme , Ostéoclastes/anatomopathologie , Ligand de RANK/génétique , Ligand de RANK/métabolisme
10.
J Clin Invest ; 131(1)2021 01 04.
Article de Anglais | MEDLINE | ID: mdl-33108352

RÉSUMÉ

Zeb1, a zinc finger E-box binding homeobox epithelial-mesenchymal transition (EMT) transcription factor, confers properties of "stemness," such as self-renewal, in cancer. Yet little is known about the function of Zeb1 in adult stem cells. Here, we used the hematopoietic system as a well-established paradigm of stem cell biology to evaluate Zeb1-mediated regulation of adult stem cells. We employed a conditional genetic approach using the Mx1-Cre system to specifically knock out (KO) Zeb1 in adult hematopoietic stem cells (HSCs) and their downstream progeny. Acute genetic deletion of Zeb1 led to rapid-onset thymic atrophy and apoptosis-driven loss of thymocytes and T cells. A profound cell-autonomous self-renewal defect and multilineage differentiation block were observed in Zeb1-KO HSCs. Loss of Zeb1 in HSCs activated transcriptional programs of deregulated HSC maintenance and multilineage differentiation genes and of cell polarity consisting of cytoskeleton-, lipid metabolism/lipid membrane-, and cell adhesion-related genes. Notably, epithelial cell adhesion molecule (EpCAM) expression was prodigiously upregulated in Zeb1-KO HSCs, which correlated with enhanced cell survival, diminished mitochondrial metabolism, ribosome biogenesis, and differentiation capacity and an activated transcriptomic signature associated with acute myeloid leukemia (AML) signaling. ZEB1 expression was downregulated in AML patients, and Zeb1 KO in the malignant counterparts of HSCs - leukemic stem cells (LSCs) - accelerated MLL-AF9- and Meis1a/Hoxa9-driven AML progression, implicating Zeb1 as a tumor suppressor in AML LSCs. Thus, Zeb1 acts as a transcriptional regulator in hematopoiesis, critically coordinating HSC self-renewal, apoptotic, and multilineage differentiation fates required to suppress leukemic potential in AML.


Sujet(s)
Cellules souches hématopoïétiques/métabolisme , Leucémie aigüe myéloïde/métabolisme , Cellules souches tumorales/métabolisme , Protéines suppresseurs de tumeurs/métabolisme , Facteur de transcription Zeb1/métabolisme , Animaux , Délétion de gène , Cellules souches hématopoïétiques/anatomopathologie , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Souris , Souris knockout , Cellules souches tumorales/anatomopathologie , Protéines suppresseurs de tumeurs/génétique , Facteur de transcription Zeb1/génétique
11.
J Clin Invest ; 130(7): 3483-3498, 2020 07 01.
Article de Anglais | MEDLINE | ID: mdl-32191640

RÉSUMÉ

The sensory nerve was recently identified as being involved in regulation of bone mass accrual. We previously discovered that prostaglandin E2 (PGE2) secreted by osteoblasts could activate sensory nerve EP4 receptor to promote bone formation by inhibiting sympathetic activity. However, the fundamental units of bone formation are active osteoblasts, which originate from mesenchymal stromal/stem cells (MSCs). Here, we found that after sensory denervation, knockout of the EP4 receptor in sensory nerves, or knockout of COX-2 in osteoblasts, could significantly promote adipogenesis and inhibit osteogenesis in adult mice. Furthermore, injection of SW033291 (a small molecule that locally increases the PGE2 level) or propranolol (a beta blocker) significantly promoted osteogenesis and inhibited adipogenesis. This effect of SW033291, but not propranolol, was abolished in conditional EP4-KO mice under normal conditions or in the bone repair process. We conclude that the PGE2/EP4 sensory nerve axis could regulate MSC differentiation in bone marrow of adult mice.


Sujet(s)
Adipogenèse , Dinoprostone/métabolisme , Cellules souches mésenchymateuses/métabolisme , Ostéogenèse , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Cellules réceptrices sensorielles/métabolisme , Animaux , Cyclooxygenase 2/métabolisme , Dinoprostone/génétique , Techniques de knock-out de gènes , Cellules souches mésenchymateuses/anatomopathologie , Souris , Souris knockout , Ostéoblastes/métabolisme , Ostéoblastes/anatomopathologie , Sous-type EP4 des récepteurs des prostaglandines E/génétique , Cellules réceptrices sensorielles/anatomopathologie
12.
JCI Insight ; 5(3)2020 02 13.
Article de Anglais | MEDLINE | ID: mdl-32051335

RÉSUMÉ

We previously established that DNA methyltransferase 3b (Dnmt3b) is the sole Dnmt responsive to fracture repair and that Dnmt3b expression is induced in progenitor cells during fracture repair. In the current study, we confirmed that Dnmt3b ablation in mesenchymal progenitor cells (MPCs) resulted in impaired endochondral ossification, delayed fracture repair, and reduced mechanical strength of the newly formed bone in Prx1-Cre;Dnmt3bf/f (Dnmt3bPrx1) mice. Mechanistically, deletion of Dnmt3b in MPCs led to reduced chondrogenic and osteogenic differentiation in vitro. We further identified Rbpjκ as a downstream target of Dnmt3b in MPCs. In fact, we located 2 Dnmt3b binding sites in the murine proximal Rbpjκ promoter and gene body and confirmed Dnmt3b interaction with the 2 binding sites by ChIP assays. Luciferase assays showed functional utilization of the Dnmt3b binding sites in murine C3H10T1/2 cells. Importantly, we showed that the MPC differentiation defect observed in Dnmt3b deficiency cells was due to the upregulation of Rbpjκ, evident by restored MPC differentiation upon Rbpjκ inhibition. Consistent with in vitro findings, Rbpjκ blockage via dual antiplatelet therapy reversed the differentiation defect and accelerated fracture repair in Dnmt3bPrx1 mice. Collectively, our data suggest that Dnmt3b suppresses Notch signaling during MPC differentiation and is necessary for normal fracture repair.


Sujet(s)
DNA (cytosine-5-)-methyltransferase/génétique , Consolidation de fracture , Fractures osseuses/physiopathologie , Récepteurs Notch/métabolisme , Régulation positive , Animaux , Fractures osseuses/métabolisme , Souris , Souris knockout , Ostéogenèse ,
13.
J Clin Invest ; 130(4): 2097-2110, 2020 04 01.
Article de Anglais | MEDLINE | ID: mdl-31961825

RÉSUMÉ

Despite the effective clinical use of steroids for the treatment of Diamond Blackfan anemia (DBA), the mechanisms through which glucocorticoids regulate human erythropoiesis remain poorly understood. We report that the sensitivity of erythroid differentiation to dexamethasone is dependent on the developmental origin of human CD34+ progenitor cells, specifically increasing the expansion of CD34+ progenitors from peripheral blood (PB) but not cord blood (CB). Dexamethasone treatment of erythroid-differentiated PB, but not CB, CD34+ progenitors resulted in the expansion of a newly defined CD34+CD36+CD71hiCD105med immature colony-forming unit-erythroid (CFU-E) population. Furthermore, proteomics analyses revealed the induction of distinct proteins in dexamethasone-treated PB and CB erythroid progenitors. Dexamethasone treatment of PB progenitors resulted in the specific upregulation of p57Kip2, a Cip/Kip cyclin-dependent kinase inhibitor, and we identified this induction as critical; shRNA-mediated downregulation of p57Kip2, but not the related p27Kip1, significantly attenuated the impact of dexamethasone on erythroid differentiation and inhibited the expansion of the immature CFU-E subset. Notably, in the context of DBA, we found that steroid resistance was associated with dysregulated p57Kip2 expression. Altogether, these data identify a unique glucocorticoid-responsive human erythroid progenitor and provide new insights into glucocorticoid-based therapeutic strategies for the treatment of patients with DBA.


Sujet(s)
Anémie de Blackfan-Diamond/métabolisme , Inhibiteur p57 de kinase cycline-dépendante/biosynthèse , Dexaméthasone/pharmacologie , Résistance aux substances/effets des médicaments et des substances chimiques , Précurseurs érythroïdes/métabolisme , Régulation positive/effets des médicaments et des substances chimiques , Adulte , Anémie de Blackfan-Diamond/traitement médicamenteux , Anémie de Blackfan-Diamond/anatomopathologie , Antigènes CD/métabolisme , Inhibiteur p27 de kinase cycline-dépendante/biosynthèse , Précurseurs érythroïdes/anatomopathologie , Femelle , Humains , Mâle
14.
J Clin Invest ; 130(4): 1843-1849, 2020 04 01.
Article de Anglais | MEDLINE | ID: mdl-31895700

RÉSUMÉ

Cancer-related anemia is present in more than 60% of newly diagnosed cancer patients and is associated with substantial morbidity and high medical costs. Drugs that enhance erythropoiesis are urgently required to decrease transfusion rates and improve quality of life. Clinical studies have observed an unexpected improvement in hemoglobin and RBC transfusion-independence in patients with acute myeloid leukemia (AML) treated with the isocitrate dehydrogenase 2 (IDH2) mutant-specific inhibitor enasidenib, leading to improved quality of life without a reduction in AML disease burden. Here, we demonstrate that enasidenib enhanced human erythroid differentiation of hematopoietic progenitors. The phenomenon was not observed with other IDH1/2 inhibitors and occurred in IDH2-deficient CRISPR-engineered progenitors independently of D-2-hydroxyglutarate. The effect of enasidenib on hematopoietic progenitors was mediated by protoporphyrin accumulation, driving heme production and erythroid differentiation in committed CD71+ progenitors rather than hematopoietic stem cells. Our results position enasidenib as a promising therapeutic agent for improvement of anemia and provide the basis for a clinical trial using enasidenib to decrease transfusion dependence in a wide array of clinical contexts.


Sujet(s)
Aminopyridines/pharmacologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cellules érythroïdes/enzymologie , Cellules souches hématopoïétiques/enzymologie , Isocitrate dehydrogenases/antagonistes et inhibiteurs , Triazines/pharmacologie , Cellules érythroïdes/cytologie , Cellules souches hématopoïétiques/cytologie , Humains , Isocitrate dehydrogenases/métabolisme , Protoporphyrines/métabolisme
15.
J Clin Invest ; 129(3): 1180-1192, 2019 03 01.
Article de Anglais | MEDLINE | ID: mdl-30620726

RÉSUMÉ

The development and function of stem and progenitor cells that produce blood cells are vital in physiology. GATA-binding protein 2 (GATA2) mutations cause GATA-2 deficiency syndrome involving immunodeficiency, myelodysplastic syndrome, and acute myeloid leukemia. GATA-2 physiological activities necessitate that it be strictly regulated, and cell type-specific enhancers fulfill this role. The +9.5 intronic enhancer harbors multiple conserved cis-elements, and germline mutations of these cis-elements are pathogenic in humans. Since mechanisms underlying how GATA2 enhancer disease mutations impact hematopoiesis and pathology are unclear, we generated mouse models of the enhancer mutations. While a multi-motif mutant was embryonically lethal, a single-nucleotide Ets motif mutant was viable, and steady-state hematopoiesis was normal. However, the Ets motif mutation abrogated stem/progenitor cell regeneration following stress. These results reveal a new mechanism in human genetics, in which a disease predisposition mutation inactivates enhancer regenerative activity, while sparing developmental activity. Mutational sensitization to stress that instigates hematopoietic failure constitutes a paradigm for GATA-2 deficiency syndrome and other contexts of GATA-2-dependent pathogenesis.


Sujet(s)
Éléments activateurs (génétique) , Déficience en GATA2 , Facteur de transcription GATA-2 , Mutation germinale , Hématopoïèse/génétique , Motifs nucléotidiques , Régénération/génétique , Animaux , Déficience en GATA2/génétique , Déficience en GATA2/métabolisme , Facteur de transcription GATA-2/génétique , Facteur de transcription GATA-2/métabolisme , Humains , Souris , Souches mutantes de souris
16.
JCI Insight ; 3(23)2018 12 06.
Article de Anglais | MEDLINE | ID: mdl-30518681

RÉSUMÉ

New techniques for single-cell analysis have led to insights into hematopoiesis and the immune system, but the ability of these techniques to cross-validate and reproducibly identify the biological variation in diverse human samples is currently unproven. We therefore performed a comprehensive assessment of human bone marrow cells using both single-cell RNA sequencing and multiparameter flow cytometry from 20 healthy adult human donors across a broad age range. These data characterize variation between healthy donors as well as age-associated changes in cell population frequencies. Direct comparison of techniques revealed discrepancy in the quantification of T lymphocyte and natural killer cell populations. Orthogonal validation of immunophenotyping using mass cytometry demonstrated a strong correlation with flow cytometry. Technical replicates using single-cell RNA sequencing matched robustly, while biological replicates showed variation. Given the increasing use of single-cell technologies in translational research, this resource serves as an important reference data set and highlights opportunities for further refinement.


Sujet(s)
Moelle osseuse/immunologie , Cytométrie en flux/méthodes , Analyse de séquence d'ARN/méthodes , Analyse sur cellule unique/méthodes , Immunité acquise , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Moelle osseuse/métabolisme , Cellules de la moelle osseuse/immunologie , Différenciation cellulaire , Femelle , Hématopoïèse , Humains , Immunophénotypage , Cellules tueuses naturelles , Mâle , Adulte d'âge moyen , Reproductibilité des résultats , Lymphocytes T , Jeune adulte
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...