Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 85
Filtrer
2.
J Thromb Haemost ; 22(1): 172-187, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37678548

RÉSUMÉ

BACKGROUND: Neutrophils participate in the pathogenesis of thrombosis through the formation of neutrophil extracellular traps (NETs). Thrombosis is the main cause of morbidity and mortality in patients with myeloproliferative neoplasms (MPNs). Recent studies have shown an increase in NET formation (NETosis) both in patients with JAK2V617F neutrophils and in mouse models, and reported the participation of NETosis in the pathophysiology of thrombosis in mice. OBJECTIVES: This study investigated whether JAK2V617F neutrophils are sufficient to promote thrombosis or whether their cooperation with other blood cell types is necessary. METHODS: NETosis was studied in PF4iCre;Jak2V617F/WT mice expressing JAK2V617F in all hematopoietic lineages, as occurs in MPNs, and in MRP8Cre;Jak2V617F/WT mice in which JAK2V617F is expressed only in leukocytes. RESULTS: In PF4iCre;Jak2V617F/WT mice, an increase in NETosis and spontaneous lung thrombosis abrogated by DNAse administration were observed. The absence of spontaneous NETosis or lung thrombosis in MRP8Cre;Jak2V617F/WT mice suggested that mutated neutrophils alone are not sufficient to induce thrombosis. Ex vivo experiments demonstrated that JAK2V617F-mutated platelets trigger NETosis by JAK2V617F-mutated neutrophils. Aspirin treatment in PF4iCre;Jak2V617F/WT mice reduced NETosis and reduced lung thrombosis. In cytoreductive-therapy-free patients with MPN treated with aspirin, plasma NET marker concentrations were lower than that in patients with MPN not treated with aspirin. CONCLUSION: Our study demonstrates that JAK2V617F neutrophils alone are not sufficient to promote thrombosis; rather, platelets cooperate with neutrophils to promote NETosis in vivo. A new role for aspirin in thrombosis prevention in MPNs was also identified.


Sujet(s)
Pièges extracellulaires , Syndromes myéloprolifératifs , Tumeurs , Thrombose , Thrombose veineuse , Humains , Souris , Animaux , Granulocytes neutrophiles/métabolisme , Pièges extracellulaires/métabolisme , Tumeurs/métabolisme , Syndromes myéloprolifératifs/génétique , Kinase Janus-2/génétique , Thrombose veineuse/métabolisme , Acide acétylsalicylique
3.
Fac Rev ; 12: 23, 2023.
Article de Anglais | MEDLINE | ID: mdl-37771602

RÉSUMÉ

Primary myelofibrosis (PMF), polycythemia vera (PV) and essential thrombocythemia (ET) form the classical BCR-ABL1-negative myeloproliferative neoplasms (MPNs) that are driven by a constitutive activation of JAK2 signaling. PMF as well as secondary MF (post-ET and post-PV MF) are the most aggressive MPNs. Presently, there is no curative treatment, except allogenic hematopoietic stem cell transplantation. JAK inhibitors, essentially ruxolitinib, are the therapy of reference for intermediate and high-risk MF. However, presently the current JAK inhibitors behave mainly as anti-inflammatory drugs, improving general symptoms and spleen size without major impact on disease progression. A better understanding of the genetics of MF, the biology of its leukemic stem cells (LSCs), the mechanisms of fibrosis and of cytopenia and the role of inflammatory cytokines has led to new approaches with the development of numerous therapeutic agents that target epigenetic regulation, telomerase, apoptosis, cell cycle, cytokines and signaling. Furthermore, the use of a new less toxic form of interferon-α has been revived, as it is presently one of the only molecules that targets the mutated clone. These new approaches have different aims: (a) to provide alternative therapy to JAK inhibition; (b) to correct cytopenia; and (c) to inhibit fibrosis development. However, the main important goal is to find new disease modifier treatments, which will profoundly modify the progression of the disease without major toxicity. Presently the most promising approaches consist of the inhibition of telomerase and the combination of JAK2 inhibitors (ruxolitinib) with either a BCL2/BCL-xL or BET inhibitor. Yet, the most straightforward future approaches can be considered to be the development of and/or selective inhibition of JAK2V617F and the targeting MPL and calreticulin mutants by immunotherapy. It can be expected that the therapy of MF will be significantly improved in the coming years.

4.
Blood Cancer J ; 13(1): 106, 2023 07 10.
Article de Anglais | MEDLINE | ID: mdl-37423955

RÉSUMÉ

The Janus kinase 2 (JAK2)-driven myeloproliferative neoplasms (MPNs) are chronic malignancies associated with high-risk complications and suboptimal responses to JAK inhibitors such as ruxolitinib. A better understanding of cellular changes induced by ruxolitinib is required to develop new combinatory therapies to improve treatment efficacy. Here, we demonstrate that ruxolitinib induced autophagy in JAK2V617F cell lines and primary MPN patient cells through the activation of protein phosphatase 2A (PP2A). Inhibition of autophagy or PP2A activity along with ruxolitinib treatment reduced proliferation and increased the death of JAK2V617F cells. Accordingly, proliferation and clonogenic potential of JAK2V617F-driven primary MPN patient cells, but not of normal hematopoietic cells, were markedly impaired by ruxolitinib treatment with autophagy or PP2A inhibitor. Finally, preventing ruxolitinib-induced autophagy with a novel potent autophagy inhibitor Lys05 improved leukemia burden reduction and significantly prolonged the mice's overall survival compared with ruxolitinib alone. This study demonstrates that PP2A-dependent autophagy mediated by JAK2 activity inhibition contributes to resistance to ruxolitinib. Altogether, our data support that targeting autophagy or its identified regulator PP2A could enhance sensitivity to ruxolitinib of JAK2V617F MPN cells and improve MPN patient care.


Sujet(s)
Syndromes myéloprolifératifs , Tumeurs , Souris , Animaux , Kinase Janus-2 , Protein Phosphatase 2/génétique , Syndromes myéloprolifératifs/traitement médicamenteux , Syndromes myéloprolifératifs/génétique , Syndromes myéloprolifératifs/métabolisme , Autophagie , Mutation
5.
Leukemia ; 37(6): 1287-1297, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-37100881

RÉSUMÉ

Heterozygous mutation targeting proline 95 in Serine/Arginine-rich Splicing Factor 2 (SRSF2) is associated with V617F mutation in Janus Activated Kinase 2 (JAK2) in some myeloproliferative neoplasms (MPNs), most commonly primary myelofibrosis. To explore the interaction of Srsf2P95H with Jak2V617F, we generated Cre-inducible knock-in mice expressing these mutants under control of the stem cell leukemia (Scl) gene promoter. In transplantation experiments, Srsf2P95H unexpectedly delayed myelofibrosis induced by Jak2V617F and decreased TGFß1 serum level. Srsf2P95H reduced the competitiveness of transplanted Jak2V617F hematopoietic stem cells while preventing their exhaustion. RNA sequencing of sorted megakaryocytes identified an increased number of splicing events when the two mutations were combined. Focusing on JAK/STAT pathway, Jak2 exon 14 skipping was promoted by Srsf2P95H, an event detected in patients with JAK2V617F and SRSF2P95 co-mutation. The skipping event generates a truncated inactive JAK2 protein. Accordingly, Srsf2P95H delays myelofibrosis induced by the thrombopoietin receptor agonist Romiplostim in Jak2 wild-type animals. These results unveil JAK2 exon 14 skipping promotion as a strategy to reduce JAK/STAT signaling in pathological conditions.


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Syndromes myéloprolifératifs , Myélofibrose primitive , Animaux , Souris , Kinase Janus-2/génétique , Janus kinases/génétique , Mutation , Syndromes myéloprolifératifs/génétique , Myélofibrose primitive/génétique , Protéines de liaison à l'ARN/génétique , Transduction du signal , Facteurs de transcription STAT/génétique
6.
Haematologica ; 108(8): 2130-2145, 2023 08 01.
Article de Anglais | MEDLINE | ID: mdl-36794499

RÉSUMÉ

Sustained ANKRD26 expression associated with germline ANKRD26 mutations causes thrombocytopenia 2 (THC2), an inherited platelet disorder associated with a predisposition to leukemia. Some patients also present with erythrocytosis and/or leukocytosis. Using multiple human-relevant in vitro models (cell lines, primary patients' cells and patient-derived induced pluripotent stem cells) we demonstrate for the first time that ANKRD26 is expressed during the early steps of erythroid, megakaryocyte and granulocyte differentiation, and is necessary for progenitor cell proliferation. As differentiation progresses, ANKRD26 expression is progressively silenced, to complete the cellular maturation of the three myeloid lineages. In primary cells, abnormal ANKRD26 expression in committed progenitors directly affects the proliferation/differentiation balance for the three cell types. We show that ANKRD26 interacts with and crucially modulates the activity of MPL, EPOR and G-CSFR, three homodimeric type I cytokine receptors that regulate blood cell production. Higher than normal levels of ANKRD26 prevent the receptor internalization that leads to increased signaling and cytokine hypersensitivity. These findings afford evidence how ANKRD26 overexpression or the absence of its silencing during differentiation is responsible for myeloid blood cell abnormalities in patients with THC2.


Sujet(s)
Leucémies , Récepteurs aux cytokines , Humains , Cytokines , Hématopoïèse , Leucémies/anatomopathologie , Différenciation cellulaire , Protéines et peptides de signalisation intercellulaire
7.
Blood ; 141(8): 917-929, 2023 02 23.
Article de Anglais | MEDLINE | ID: mdl-36356299

RÉSUMÉ

Mutant calreticulin (CALR) proteins resulting from a -1/+2 frameshifting mutation of the CALR exon 9 carry a novel C-terminal amino acid sequence and drive the development of myeloproliferative neoplasms (MPNs). Mutant CALRs were shown to interact with and activate the thrombopoietin receptor (TpoR/MPL) in the same cell. We report that mutant CALR proteins are secreted and can be found in patient plasma at levels up to 160 ng/mL, with a mean of 25.64 ng/mL. Plasma mutant CALR is found in complex with soluble transferrin receptor 1 (sTFR1) that acts as a carrier protein and increases mutant CALR half-life. Recombinant mutant CALR proteins bound and activated the TpoR in cell lines and primary megakaryocytic progenitors from patients with mutated CALR in which they drive thrombopoietin-independent colony formation. Importantly, the CALR-sTFR1 complex remains functional for TpoR activation. By bioluminescence resonance energy transfer assay, we show that mutant CALR proteins produced in 1 cell can specifically interact in trans with the TpoR on a target cell. In comparison with cells that only carry TpoR, cells that carry both TpoR and mutant CALR are hypersensitive to exogenous mutant CALR proteins and respond to levels of mutant CALR proteins similar to those in patient plasma. This is consistent with CALR-mutated cells that expose TpoR carrying immature N-linked sugars at the cell surface. Thus, secreted mutant CALR proteins will act more specifically on the MPN clone. In conclusion, a chaperone, CALR, can turn into a rogue cytokine through somatic mutation of its encoding gene.


Sujet(s)
Syndromes myéloprolifératifs , Tumeurs , Humains , Cytokines/métabolisme , Calréticuline/génétique , Syndromes myéloprolifératifs/génétique , Mutation , Facteurs immunologiques , Kinase Janus-2/génétique
8.
Br J Haematol ; 198(1): 131-136, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35355248

RÉSUMÉ

Myeloproliferative neoplasms (MPN) are mainly sporadic but inherited variants have been associated with higher risk development. Here, we identified an EPOR variant (EPORP488S ) in a large family diagnosed with JAK2V617F -positive polycythaemia vera (PV) or essential thrombocytosis (ET). We investigated its functional impact on JAK2V617F clonal amplification in patients and found that the variant allele fraction (VAF) was low in PV progenitors but increase strongly in mature cells. Moreover, we observed that EPORP488S alone induced a constitutive phosphorylation of STAT5 in cell lines or primary cells. Overall, this study points for searching inherited-risk alleles affecting the JAK2/STAT pathway in MPN.


Sujet(s)
Syndromes myéloprolifératifs , Polyglobulie primitive essentielle , Récepteur érythropoïétine , Thrombocytémie essentielle , Allèles , Mutation gain de fonction , Humains , Kinase Janus-2/génétique , Kinase Janus-2/métabolisme , Mutation , Syndromes myéloprolifératifs/diagnostic , Syndromes myéloprolifératifs/génétique , Polyglobulie primitive essentielle/génétique , Récepteur érythropoïétine/génétique , Thrombocytémie essentielle/génétique
9.
Int Rev Cell Mol Biol ; 366: 125-185, 2022.
Article de Anglais | MEDLINE | ID: mdl-35153003

RÉSUMÉ

Over the past decades, a variety of MPN mouse models have been developed to express in HSC the main mutations identified in patients: JAK2V617F, CALRdel52 or ins5 and MPLW515L. These models mimic quite faithfully human PV or ET with their natural evolutions into MF and their hemostasis complications, demonstrating the driver function of these mutations in MPN. Here, we review these models and show how they have improved our general understanding of MPN regarding (1) the mechanisms of fibrosis, thrombosis/hemorrhages and disease initiation, (2) the roles of additional mutations and signaling pathways in disease progression and (3) the preclinical development of novel therapies. We also address controversial results between these models and remind how these models may differ from human MPN onset and also how basically mice are not humans, encouraging caution when one draw lessons from mice to humans. Furthermore, the contribution of germline genetic predisposition, HSC and niche aging, metabolic, oxidative, replicative or genotoxic stress, inflammation, immune escape and additional mutations need to be considered in further investigations to encompass the full complexity of human MPN in mice.


Sujet(s)
Thrombocytémie essentielle , Animaux , Modèles animaux de maladie humaine , Évolution de la maladie , Prédisposition génétique à une maladie , Humains , Souris , Mutation , Récepteurs à la thrombopoïétine/génétique , Thrombocytémie essentielle/génétique
10.
Bone ; 153: 116167, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34492359

RÉSUMÉ

Several drugs are able to reduce fracture risk in osteoporotic patients. Incident fractures occur despite good adherence to treatment. Inadequate response has been found related to high serum bone biomarkers of bone turnover. We here aimed to analyze bone microarchitecture and cellular profiles of inadequate responders. We retrospectively analyzed bone biopsies from patients with major fractures despite long-term treatment (inadequate responder [IR] n = 31) in comparison to patients with untreated osteoporosis (U-OP, n = 31) and controls without osteoporosis (Ctrl, n = 16). Bone samples were analyzed by histomorphometry and micro-computed tomography. Clinical and bone turnover markers and bone mineral density were assessed. As compared with U-OP patients, IRs were older (mean age 69.7 ± 8.8 vs 63.3 ± 9.3 years, p = 0.007) and had lower mean hip bone mineral density (0.685 ± 0.116 vs 0.786 ± 0.093 g/cm2), p = 0.019 and T-score (-2.3 ± 0.769 vs -1.6 ± 0.900, p = 0.032). BV/TV was lower for IRs than U-OP patients and Ctrls (13.9 ± 3.8% vs 15.2 ± 5.1 and 17.6 ± 5.2%, p = 0.044) as was trabecular thickness (145.6 ± 23.1 vs 160.5 ± 22.7 and 153.7 ± 21.4 µm, p = 0.033). Mean structure model index was lower for IRs than U-OP patients (1.9 ± 0.806 vs 2.4 ± 0.687, p = 0.042) and osteoclast number was higher for IRs than U-OP patients and Ctrls (0.721 ± 0.611 vs 0.394 ± 0.393 and 0.199 ± 0.071 mm-2, p < 0.001). The mean Obl.S/BS was lower for IRs than U-OP patients and Ctrls (1.2 ± 1.3 vs 1.9 ± 1.4 and 3.0 ± 0.638 mm-2, p < 0.0001), and the mean number of labelled surfaces was lower for IRs than U-OP patients (51.6% vs 87%, p = 0.002). Cortical parameters did not significantly differ. We show an imbalance of bone remodeling in favor of bone resorption in IRs. The persistence of high bone resorption suggests insufficient inhibition of bone resorption that could explain the incident fractures with anti-osteoporotic drug use. Adaptation to treatment should be considered to inhibit bone resorption and prevent further fractures.


Sujet(s)
Résorption osseuse , Ostéoporose , Fractures ostéoporotiques , Sujet âgé , Densité osseuse , Humains , Adulte d'âge moyen , Ostéoclastes , Ostéoporose/traitement médicamenteux , Études rétrospectives , Microtomographie aux rayons X
11.
Blood ; 138(22): 2231-2243, 2021 12 02.
Article de Anglais | MEDLINE | ID: mdl-34407546

RÉSUMÉ

Classical BCR-ABL-negative myeloproliferative neoplasms (MPNs) are clonal disorders of hematopoietic stem cells (HSCs) caused mainly by recurrent mutations in genes encoding JAK2 (JAK2), calreticulin (CALR), or the thrombopoietin receptor (MPL). Interferon α (IFNα) has demonstrated some efficacy in inducing molecular remission in MPNs. To determine factors that influence molecular response rate, we evaluated the long-term molecular efficacy of IFNα in patients with MPN by monitoring the fate of cells carrying driver mutations in a prospective observational and longitudinal study of 48 patients over more than 5 years. We measured the clonal architecture of early and late hematopoietic progenitors (84 845 measurements) and the global variant allele frequency in mature cells (409 measurements) several times per year. Using mathematical modeling and hierarchical Bayesian inference, we further inferred the dynamics of IFNα-targeted mutated HSCs. Our data support the hypothesis that IFNα targets JAK2V617F HSCs by inducing their exit from quiescence and differentiation into progenitors. Our observations indicate that treatment efficacy is higher in homozygous than heterozygous JAK2V617F HSCs and increases with high IFNα dose in heterozygous JAK2V617F HSCs. We also found that the molecular responses of CALRm HSCs to IFNα were heterogeneous, varying between type 1 and type 2 CALRm, and a high dose of IFNα correlates with worse outcomes. Our work indicates that the long-term molecular efficacy of IFNα implies an HSC exhaustion mechanism and depends on both the driver mutation type and IFNα dose.


Sujet(s)
Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Facteurs immunologiques/usage thérapeutique , Interféron alpha/usage thérapeutique , Mutation/effets des médicaments et des substances chimiques , Syndromes myéloprolifératifs/traitement médicamenteux , Calréticuline/génétique , Cellules souches hématopoïétiques/métabolisme , Cellules souches hématopoïétiques/anatomopathologie , Humains , Facteurs immunologiques/pharmacologie , Interféron alpha/pharmacologie , Kinase Janus-2/génétique , Études longitudinales , Syndromes myéloprolifératifs/génétique , Syndromes myéloprolifératifs/anatomopathologie , Études prospectives , Récepteurs à la thrombopoïétine/génétique , Cellules cancéreuses en culture
12.
Hemasphere ; 5(7): e593, 2021 Jul.
Article de Anglais | MEDLINE | ID: mdl-34131633

RÉSUMÉ

Mutations in the calreticulin (CALR) gene are seen in about 30% of essential thrombocythemia and primary myelofibrosis patients. To address the contribution of the human CALR mutants to the pathogenesis of myeloproliferative neoplasms (MPNs) in an endogenous context, we modeled the CALRdel52 and CALRins5 mutants by induced pluripotent stem cell (iPSC) technology using CD34+ progenitors from 4 patients. We describe here the generation of several clones of iPSC carrying heterozygous CALRdel52 or CALRins5 mutations. We showed that CALRdel52 induces a stronger increase in progenitors than CALRins5 and that both CALRdel52 and CALRins5 mutants favor an expansion of the megakaryocytic lineage. Moreover, we found that both CALRdel52 and CALRins5 mutants rendered colony forming unit-megakaryocyte (CFU-MK) independent from thrombopoietin (TPO), and promoted a mild constitutive activation level of signal transducer and activator of transcription 3 in megakaryocytes. Unexpectedly, a mild increase in the sensitivity of colony forming unit-granulocyte (CFU-G) to granulocyte-colony stimulating factor was also observed in iPSC CALRdel52 and CALRins5 compared with control iPSC. Moreover, CALRdel52-induced megakaryocytic spontaneous growth is more dependent on Janus kinase 2/phosphoinositide 3-kinase/extracellular signal-regulated kinase than TPO-mediated growth and opens a therapeutic window for treatments in CALR-mutated MPN. The iPSC models described here represent an interesting platform for testing newly developed inhibitors. Altogether, this study shows that CALR-mutated iPSC recapitulate MPN phenotypes in vitro and may be used for drug screening.

13.
Blood ; 138(6): 480-485, 2021 08 12.
Article de Anglais | MEDLINE | ID: mdl-34010413

RÉSUMÉ

Congenital amegakaryocytic thrombocytopenia (CAMT) is a severe inherited thrombocytopenia due to loss-of-function mutations affecting the thrombopoietin (TPO) receptor, MPL. Here, we report a new homozygous MPL variant responsible for CAMT in 1 consanguineous family. The propositus and her sister presented with severe thrombocytopenia associated with mild anemia. Next-generation sequencing revealed the presence of a homozygous MPLR464G mutation resulting in a weak cell-surface expression of the receptor in platelets. In cell lines, we observed a defect in MPLR464G maturation associated with its retention in the endoplasmic reticulum. The low cell-surface expression of MPLR464G induced very limited signaling with TPO stimulation, leading to survival and reduced proliferation of cells. Overexpression of a myeloproliferative neoplasm-associated calreticulin (CALR) mutant did not rescue trafficking of MPLR464G to the cell surface and did not induce constitutive signaling. However, it unexpectedly restored a normal response to eltrombopag (ELT), but not to TPO. This effect was only partially mimicked by the purified recombinant CALR mutant protein. Finally, the endogenous CALR mutant was able to restore the megakaryocyte differentiation of patient CD34+ cells carrying MPLR464G in response to ELT.


Sujet(s)
Benzoates/pharmacologie , Calréticuline , Insuffisances médullaires congénitales , Hydrazines/pharmacologie , Mutation faux-sens , Pyrazoles/pharmacologie , Récepteurs à la thrombopoïétine , Thrombopénie , Adulte , Substitution d'acide aminé , Calréticuline/génétique , Calréticuline/métabolisme , Enfant , Enfant d'âge préscolaire , Insuffisances médullaires congénitales/traitement médicamenteux , Insuffisances médullaires congénitales/génétique , Insuffisances médullaires congénitales/métabolisme , Insuffisances médullaires congénitales/anatomopathologie , Femelle , Cellules HEK293 , Homozygote , Humains , Nourrisson , Mâle , Récepteurs à la thrombopoïétine/génétique , Récepteurs à la thrombopoïétine/métabolisme , Thrombopénie/traitement médicamenteux , Thrombopénie/génétique , Thrombopénie/métabolisme , Thrombopénie/anatomopathologie
14.
Elife ; 102021 05 18.
Article de Anglais | MEDLINE | ID: mdl-34002698

RÉSUMÉ

High-throughput single-cell methods have uncovered substantial heterogeneity in the pool of hematopoietic stem and progenitor cells (HSPCs), but how much instruction is inherited by offspring from their heterogeneous ancestors remains unanswered. Using a method that enables simultaneous determination of common ancestor, division number, and differentiation status of a large collection of single cells, our data revealed that murine cells that derived from a common ancestor had significant similarities in their division progression and differentiation outcomes. Although each family diversifies, the overall collection of cell types observed is composed of homogeneous families. Heterogeneity between families could be explained, in part, by differences in ancestral expression of cell surface markers. Our analyses demonstrate that fate decisions of cells are largely inherited from ancestor cells, indicating the importance of common ancestor effects. These results may have ramifications for bone marrow transplantation and leukemia, where substantial heterogeneity in HSPC behavior is observed.


Sujet(s)
Différenciation cellulaire , Prolifération cellulaire , Cellules souches hématopoïétiques/physiologie , Animaux , Moelle osseuse , Cellules de la moelle osseuse , Cellules cultivées , Cellules souches hématopoïétiques/classification , Souris , Souris de lignée C57BL
15.
J Clin Invest ; 131(11)2021 06 01.
Article de Anglais | MEDLINE | ID: mdl-33914703

RÉSUMÉ

Myelofibrosis (MF) is a non-BCR-ABL myeloproliferative neoplasm associated with poor outcomes. Current treatment has little effect on the natural history of the disease. MF results from complex interactions between (a) the malignant clone, (b) an inflammatory context, and (c) remodeling of the bone marrow (BM) microenvironment. Each of these points is a potential target of PPARγ activation. Here, we demonstrated the therapeutic potential of PPARγ agonists in resolving MF in 3 mouse models. We showed that PPARγ agonists reduce myeloproliferation, modulate inflammation, and protect the BM stroma in vitro and ex vivo. Activation of PPARγ constitutes a relevant therapeutic target in MF, and our data support the possibility of using PPARγ agonists in clinical practice.


Sujet(s)
Antinéoplasiques/pharmacologie , Tumeurs hématologiques/traitement médicamenteux , Protéines tumorales/agonistes , Tumeurs expérimentales/traitement médicamenteux , Récepteur PPAR gamma/agonistes , Myélofibrose primitive/traitement médicamenteux , Animaux , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/anatomopathologie , Modèles animaux de maladie humaine , Tumeurs hématologiques/génétique , Tumeurs hématologiques/métabolisme , Tumeurs hématologiques/anatomopathologie , Souris , Souris transgéniques , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Tumeurs expérimentales/génétique , Tumeurs expérimentales/métabolisme , Tumeurs expérimentales/anatomopathologie , Récepteur PPAR gamma/génétique , Récepteur PPAR gamma/métabolisme , Myélofibrose primitive/génétique , Myélofibrose primitive/métabolisme , Myélofibrose primitive/anatomopathologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/génétique
16.
Arthritis Rheumatol ; 73(2): 324-329, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-32909692

RÉSUMÉ

OBJECTIVE: The coexistence of calcium pyrophosphate dihydrate (CPPD) and monosodium urate monohydrate crystals in gouty tophi has rarely been reported. We undertook this study to investigate CPPD crystal deposits in a series of surgically removed gouty tophi and to identify factors associated with these deposits. METHODS: Twenty-five tophi from 22 gout patients were analyzed using polarized light microscopy, field emission scanning electron microscopy (FESEM), and µ Fourier transform infrared (µFTIR) spectroscopy. RESULTS: Tophi consisted of multiple lobules separated by fibrous septa and surrounded by a foreign-body giant cell reaction. CPPD crystal aggregates were identified in 9 of 25 tophi from 6 patients. CPPD crystals were dispersed or highly compacted, localized at the edge or inside the tophus lobules, with some lobules completely filled with crystals. Both monoclinic and triclinic CPPD crystal phases were identified using FESEM and µFTIR. Compared to patients without CPPD, those with CPPD-containing tophi were older (mean 60.5 years versus 47.2 years; P = 0.009), and had longer-term gout duration (mean 17.0 years versus mean 9.0 years; P < 0.05) and tophi duration (mean 10.0 years versus mean 4.6 years; P < 0.01). None of the patients had radiographic chondrocalcinosis of the knee or wrist. CONCLUSION: CPPD crystal formation seems to be a late and frequent event of tophus maturation, occurring more frequently with aging, and could contribute to the speed of tophus dissolution and the apparent persistence of tophus sometimes observed even after effective, long-lasting urate-lowering therapy.


Sujet(s)
Diphosphate de calcium/analyse , Goutte/anatomopathologie , Acide urique/analyse , Adulte , Facteurs âges , Sujet âgé , Humains , Mâle , Microscopie électronique à balayage , Microscopie en lumière polarisée , Adulte d'âge moyen , Spectroscopie infrarouge à transformée de Fourier , Facteurs temps
18.
Am J Nephrol ; 51(10): 833-838, 2020.
Article de Anglais | MEDLINE | ID: mdl-32911468

RÉSUMÉ

BACKGROUND: The incidence of skeletal fractures is high in dialysis patients. Current available tools are insufficient to predict bone fragility. We analyzed the microarchitecture in patients on dialysis therapy using bone biopsies and peripheral microcomputed tomography. METHODS: We analyzed 12 trans-iliac bone biopsies of patients with recent fractures. Bone microarchitecture was assessed in the bone cores by histology (2D-), microcomputed tomography (3D-µCT), and high-resolution peripheral quantitative computed tomography (HR-pQCT) at the tibia. RESULTS: Trabecular bone volume/tissue volume was similar in 2D histology and 3D-µCT (p = 0.40), while lower in HR-pQCT (p < 0.01). There was no correlation in trabecular microarchitectural indices between 2-histology and 3D-µCT, or HR-pQCT. The 3D-µCT cortical thickness (Ct.Th) were positively correlated with 2D (p < 0.05), but with HR-pQCT (p = 0.33). Ct.Th was lower in patients with ≥2 vertebral fractures than with one fracture. CONCLUSIONS: 3D-µCT is a reliable method for the measurement of cortical bone in bone biopsies. Prospective studies are awaited to address its value in discriminating fracture risk.


Sujet(s)
Os cortical/imagerie diagnostique , Défaillance rénale chronique/complications , Fractures ostéoporotiques/épidémiologie , Dialyse rénale/effets indésirables , Microtomographie aux rayons X , Sujet âgé , Sujet âgé de 80 ans ou plus , Biopsie , Os cortical/anatomopathologie , Femelle , Études de suivi , Humains , Incidence , Défaillance rénale chronique/thérapie , Mâle , Adulte d'âge moyen , Fractures ostéoporotiques/étiologie , Fractures ostéoporotiques/anatomopathologie , Études prospectives , Reproductibilité des résultats , Appréciation des risques/méthodes
19.
Nat Commun ; 11(1): 4886, 2020 09 28.
Article de Anglais | MEDLINE | ID: mdl-32985500

RÉSUMÉ

Somatic mutations in the calreticulin (CALR) gene are associated with approximately 30% of essential thrombocythemia (ET) and primary myelofibrosis (PMF). CALR mutations, including the two most frequent 52 bp deletion (del52) and 5 bp insertion (ins5), induce a frameshift to the same alternative reading frame generating new C-terminal tails. In patients, del52 and ins5 induce two phenotypically distinct myeloproliferative neoplasms (MPNs). They are equally found in ET, but del52 is more frequent in PMF. We generated heterozygous and homozygous conditional inducible knock-in (KI) mice expressing a chimeric murine CALR del52 or ins5 with the human mutated C-terminal tail to investigate their pathogenic effects on hematopoiesis. Del52 induces greater phenotypic changes than ins5 including thrombocytosis, leukocytosis, splenomegaly, bone marrow hypocellularity, megakaryocytic lineage amplification, expansion and competitive advantage of the hematopoietic stem cell compartment. Homozygosity amplifies these features, suggesting a distinct contribution of homozygous clones to human MPNs. Moreover, homozygous del52 KI mice display features of a penetrant myelofibrosis-like disorder with extramedullary hematopoiesis linked to splenomegaly, megakaryocyte hyperplasia and the presence of reticulin fibers. Overall, modeling del52 and ins5 mutations in mice successfully recapitulates the differences in phenotypes observed in patients.


Sujet(s)
Calréticuline/génétique , Myélofibrose primitive/génétique , Thrombocytémie essentielle/génétique , Animaux , Calréticuline/métabolisme , Modèles animaux de maladie humaine , Femelle , Cellules souches hématopoïétiques/métabolisme , Homozygote , Humains , Kinase Janus-2/génétique , Kinase Janus-2/métabolisme , Mâle , Souris , Souris de lignée C57BL , Mutagenèse par insertion , Phénotype , Myélofibrose primitive/métabolisme , Délétion de séquence , Thrombocytémie essentielle/métabolisme
20.
Oncogene ; 39(31): 5323-5337, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32572159

RÉSUMÉ

Mutations of calreticulin (CALRm) define a subtype of myeloproliferative neoplasms (MPN). We studied the biological and genetic features of CALR-mutated essential thrombocythemia and myelofibrosis patients. In most cases, CALRm were found in granulocytes, monocytes, B and NK cells, but also in T cells. However, the type 1 CALRm spreads more easily than the type 2 CALRm in lymphoid cells. The CALRm were also associated with an early clonal dominance at the level of hematopoietic stem and progenitor cells (HSPC) with no significant increase during granulo/monocytic differentiation in most cases. Moreover, we found that half of type 2 CALRm patients harbors some homozygous progenitors. Those patients were associated with a higher clonal dominance during granulo/monocytic differentiation than patients with only heterozygous type 2 CALRm progenitors. When associated mutations were present, CALRm were the first genetic event suggesting that they are both the initiating and phenotypic event. In blood, type 1 CALRm led to a greater increased number of all types of progenitors compared with the type 2 CALRm. However, both types of CALRm induced an increase in megakaryocytic progenitors associated with a ruxolitinib-sensitive independent growth and with a mild constitutive signaling in megakaryocytes. At the transcriptional level, type 1 CALRm seems to deregulate more pathways than the type 2 CALRm in megakaryocytes. Altogether, our results show that CALRm modify both the HSPC and megakaryocyte biology with a stronger effect for type 1 than for type 2 CALRm.


Sujet(s)
Calréticuline/effets indésirables , Hématopoïèse/effets des médicaments et des substances chimiques , Syndromes myéloprolifératifs/génétique , Femelle , Humains , Mâle , Mutation
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...