Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 50
Filtrer
2.
STAR Protoc ; 5(1): 102855, 2024 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-38300798

RÉSUMÉ

RNA-binding proteins (RBPs) regulate gene expression both co-transcriptionally and post-transcriptionally. Here, we provide a protocol for photoactivatable ribonucleoside-enhanced crosslinking and immunoprecipitation followed by next-generation sequencing (PAR-CLIP-seq). PAR-CLIP-seq is a transcriptome-scale technique for identifying in vivo binding sites of RBPs at the single-nucleotide level. We detail procedures for the establishment of FLAG-RBM33 stable cell line, the sequencing library preparation, and the data analysis.


Sujet(s)
Séquençage après immunoprécipitation de la chromatine , Protéines de liaison à l'ARN , Humains , Cellules HEK293 , Sites de fixation , Protéines de liaison à l'ARN/métabolisme , Transcriptome
3.
Genes Dis ; 11(1): 382-396, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37588203

RÉSUMÉ

As the most common internal modification of mRNA, N6-methyladenosine (m6A) and its regulators modulate gene expression and play critical roles in various biological and pathological processes including tumorigenesis. It was reported previously that m6A methyltransferase (writer), methyltransferase-like 3 (METTL3) adds m6A in primary microRNAs (pri-miRNAs) and facilitates its processing into precursor miRNAs (pre-miRNAs). However, it is unknown whether m6A modification also plays a role in the maturation process of pre-miRNAs and (if so) whether such a function contributes to tumorigenesis. Here, we found that YTHDF2 is aberrantly overexpressed in acute myeloid leukemia (AML) patients, especially in relapsed patients, and plays an oncogenic role in AML. Moreover, YTHDF2 promotes expression of miR-126-3p (also known as miR-126, as it is the main product of precursor miR-126 (pre-miR-126)), a miRNA that was reported as an oncomiRNA in AML, through facilitating the processing of pre-miR-126 into mature miR-126. Mechanistically, YTHDF2 recognizes m6A modification in pre-miR-126 and recruits AGO2, a regulator of pre-miRNA processing, to promote the maturation of pre-miR-126. YTHDF2 positively and negatively correlates with miR-126 and miR-126's downstream target genes, respectively, in AML patients, and forced expression of miR-126 could largely rescue YTHDF2/Ythdf2 depletion-mediated suppression on AML cell growth/proliferation and leukemogenesis, indicating that miR-126 is a functionally important target of YTHDF2 in AML. Overall, our studies not only reveal a previously unappreciated YTHDF2/miR-126 axis in AML and highlight the therapeutic potential of targeting this axis for AML treatment, but also suggest that m6A plays a role in pre-miRNA processing that contributes to tumorigenesis.

4.
Leukemia ; 37(11): 2261-2275, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37670087

RÉSUMÉ

The highly conserved MicroRNA-9 (miR-9) family consists of three members. We discovered that miR-9-1 deletion reduced mature miR-9 expression, causing 43% of the mice to display smaller size and postweaning lethality. MiR-9-1-deficient mice with growth defects experienced severe lymphopenia, but other blood cells were unaffected. The lymphopenia wasn't due to defects in hematopoietic progenitors, as mutant bone marrow (BM) cells underwent normal lymphopoiesis after transplantation into wild-type recipients. Additionally, miR-9-1-deficient mice exhibited impaired osteoblastic bone formation, as mutant mesenchymal stem cells (MSCs) failed to differentiate into osteoblastic cells (OBs). RNA sequencing revealed reduced expression of master transcription factors for osteoblastic differentiation, Runt-related transcription factor 2 (Runx2) and Osterix (Osx), and genes related to collagen formation, extracellular matrix organization, and cell adhesion, in miR-9-1-deficient MSCs. Follistatin (Fst), an antagonist of bone morphogenetic proteins (BMPs), was found to be a direct target of miR-9-1. Its deficiency led to the up-regulation of Fst, inhibiting BMP signaling in MSCs, and reducing IL-7 and IGF-1. Thus, miR-9-1 controls osteoblastic regulation of lymphopoiesis by targeting the Fst/BMP/Smad signaling axis.


Sujet(s)
Lymphopénie , microARN , Animaux , Souris , Lymphopoïèse/génétique , Protéines morphogénétiques osseuses/métabolisme , Différenciation cellulaire , microARN/génétique , microARN/métabolisme , Ostéogenèse/génétique , Ostéoblastes/métabolisme
5.
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
6.
J Clin Invest ; 133(12)2023 06 15.
Article de Anglais | MEDLINE | ID: mdl-37317974

RÉSUMÉ

Palmitoylation is a critical posttranslational modification that enables the cellular membrane localization and subsequent activation of RAS proteins, including HRAS, KRAS, and NRAS. However, the molecular mechanism that regulates RAS palmitoylation in malignant diseases remains unclear. In this issue of the JCI, Ren, Xing, and authors shed light on this topic and revealed how upregulation of RAB27B, as a consequence of CBL loss and Janus kinase 2 (JAK2) activation, contributes to leukemogenesis. The authors found that RAB27B mediated NRAS palmitoylation and plasma membrane localization by recruiting ZDHHC9. The findings suggest that targeting RAB27B could provide a promising therapeutic strategy for NRAS-driven cancers.


Sujet(s)
Tumeurs hématologiques , Leucémies , Humains , Lipoylation , Tumeurs hématologiques/génétique , Membrane cellulaire , Carcinogenèse
7.
Mol Cell ; 83(12): 2003-2019.e6, 2023 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-37257451

RÉSUMÉ

Regulation of RNA substrate selectivity of m6A demethylase ALKBH5 remains elusive. Here, we identify RNA-binding motif protein 33 (RBM33) as a previously unrecognized m6A-binding protein that plays a critical role in ALKBH5-mediated mRNA m6A demethylation of a subset of mRNA transcripts by forming a complex with ALKBH5. RBM33 recruits ALKBH5 to its m6A-marked substrate and activates ALKBH5 demethylase activity through the removal of its SUMOylation. We further demonstrate that RBM33 is critical for the tumorigenesis of head-neck squamous cell carcinoma (HNSCC). RBM33 promotes autophagy by recruiting ALKBH5 to demethylate and stabilize DDIT4 mRNA, which is responsible for the oncogenic function of RBM33 in HNSCC cells. Altogether, our study uncovers the mechanism of selectively demethylate m6A methylation of a subset of transcripts during tumorigenesis that may explain demethylation selectivity in other cellular processes, and we showed its importance in the maintenance of tumorigenesis of HNSCC.


Sujet(s)
AlkB Homolog 5, RNA demethylase , Tumeurs de la tête et du cou , Humains , Carcinome épidermoïde de la tête et du cou/génétique , AlkB Homolog 5, RNA demethylase/génétique , AlkB Homolog 5, RNA demethylase/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Protéines de liaison à l'ARN/génétique , Carcinogenèse
8.
Sci Transl Med ; 15(689): eabq8513, 2023 03 29.
Article de Anglais | MEDLINE | ID: mdl-36989375

RÉSUMÉ

Although the overall survival rate of B cell acute lymphoblastic leukemia (B-ALL) in childhood is more than 80%, it is merely 30% in refractory/relapsed and adult patients with B-ALL. This demonstrates a need for improved therapy targeting this subgroup of B-ALL. Here, we show that the ten-eleven translocation 1 (TET1) protein, a dioxygenase involved in DNA demethylation, is overexpressed and plays a crucial oncogenic role independent of its catalytic activity in B-ALL. Consistent with its oncogenic role in B-ALL, overexpression of TET1 alone in normal precursor B cells is sufficient to transform the cells and cause B-ALL in mice within 3 to 4 months. We found that TET1 protein is stabilized and overexpressed because of its phosphorylation mediated by protein kinase C epsilon (PRKCE) and ATM serine/threonine kinase (ATM), which are also overexpressed in B-ALL. Mechanistically, TET1 recruits STAT5B to the promoters of CD72 and JCHAIN and promotes their transcription, which in turn promotes B-ALL development. Destabilization of TET1 protein by treatment with PKC or ATM inhibitors (staurosporine or AZD0156; both tested in clinical trials), or by pharmacological targeting of STAT5B, greatly decreases B-ALL cell viability and inhibits B-ALL progression in vitro and in vivo. The combination of AZD0156 with staurosporine or vincristine exhibits a synergistic effect on inhibition of refractory/relapsed B-ALL cell survival and leukemia progression in PDX models. Collectively, our study reveals an oncogenic role of the phosphorylated TET1 protein in B-ALL independent of its catalytic activity and highlights the therapeutic potential of targeting TET1 signaling for the treatment of refractory/relapsed B-ALL.


Sujet(s)
Leucémie-lymphome lymphoblastique à précurseurs B et T , Protéines proto-oncogènes , Animaux , Souris , Protéines proto-oncogènes/métabolisme , Phosphorylation , Staurosporine , Transduction du signal , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Protéines de liaison à l'ADN/métabolisme
9.
J Biol Chem ; 298(9): 102385, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-35985424

RÉSUMÉ

Asparagine synthetase (ASNS) catalyzes synthesis of asparagine (Asn) and Glu from Asp and Gln in an ATP-dependent reaction. Asparagine synthetase deficiency (ASNSD) results from biallelic mutations in the ASNS gene. Affected children exhibit congenital microcephaly, continued brain atrophy, seizures, and often premature mortality. However, the underlying mechanisms are unclear. This report describes a compound heterozygotic ASNSD child with two novel mutations in the ASNS gene, c.1118G>T (paternal) and c.1556G>A (maternal), that lead to G373V or R519H ASNS variants. Structural mapping suggested that neither variant participates directly in catalysis. Growth of cultured fibroblasts from either parent was unaffected in Asn-free medium, whereas growth of the child's cells was suppressed by about 50%. Analysis of Asn levels unexpectedly revealed that extracellular rather than intracellular Asn correlated with the reduced proliferation during incubation of the child's cells in Asn-free medium. Our attempts to ectopically express the G373V variant in either HEK293T or JRS cells resulted in minimal protein production, suggesting instability. Protein expression and purification from HEK293T cells revealed reduced activity for the R519H variant relative to WT ASNS. Expression of WT ASNS in ASNS-null JRS cells resulted in nearly complete rescue of growth in Asn-free medium, whereas we observed no proliferation for the cells expressing either the G373V or R519H variant. These results support the conclusion that the coexpression of the G373V and R519H ASNS variants leads to significantly reduced Asn synthesis, which negatively impacts cellular growth. These observations are consistent with the ASNSD phenotype.


Sujet(s)
Aminoacidopathies congénitales , Aspartate-ammonia ligase , Déficience intellectuelle , Microcéphalie , Maladies neurodégénératives , Adénosine triphosphate , Asparagine/génétique , Aspartate-ammonia ligase/composition chimique , Atrophie , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/génétique , Enfant , Cellules HEK293 , Humains , Déficience intellectuelle/génétique , Microcéphalie/génétique , Mutation
11.
Stem Cell Rev Rep ; 17(6): 2304-2313, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34561772

RÉSUMÉ

Polycomb group protein Bmi1 is essential for hematopoietic stem cell (HSC) self-renewal and terminal differentiation. However, its target genes in hematopoietic stem and progenitor cells are largely unknown. We performed gene expression profiling assays and found that genes of the Wnt signaling pathway are significantly elevated in Bmi1 null hematopoietic stem and progenitor cells (HSPCs). Bmi1 is associated with several genes of the Wnt signaling pathway in hematopoietic cells. Further, we found that Bmi1 represses Wnt gene expression in HSPCs. Importantly, loss of ß-catenin, which reduces Wnt activation, partially rescues the HSC self-renewal and differentiation defects seen in the Bmi1 null mice. Thus, we have identified Bmi1 as a novel regulator of Wnt signaling pathway in HSPCs. Given that Wnt signaling pathway plays an important role in hematopoiesis, our studies suggest that modulating Wnt signaling may hold potential for enhancing HSC self-renewal, thereby improving the outcomes of HSC transplantation.


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Voie de signalisation Wnt , Animaux , Hématopoïèse/génétique , Cellules souches hématopoïétiques , Souris , Complexe répresseur Polycomb-1/génétique , Complexe répresseur Polycomb-1/métabolisme , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Voie de signalisation Wnt/génétique
12.
Blood ; 138(26): 2838-2852, 2021 12 30.
Article de Anglais | MEDLINE | ID: mdl-34255814

RÉSUMÉ

YTHDC1 has distinct functions as a nuclear N6-methyladenosine (m6A) reader in regulating RNA metabolism. Here we show that YTHDC1 is overexpressed in acute myeloid leukemia (AML) and that it is required for the proliferation and survival of human AML cells. Genetic deletion of Ythdc1 markedly blocks AML development and maintenance as well as self-renewal of leukemia stem cells (LSCs) in vivo in mice. We found that Ythdc1 is also required for normal hematopoiesis and hematopoietic stem and progenitor cell (HSPC) maintenance in vivo. Notably, Ythdc1 haploinsufficiency reduces self-renewal of LSCs but not HSPCs in vivo. YTHDC1 knockdown has a strong inhibitory effect on proliferation of primary AML cells. Mechanistically, YTHDC1 regulates leukemogenesis through MCM4, which is a critical regulator of DNA replication. Our study provides compelling evidence that shows an oncogenic role and a distinct mechanism of YTHDC1 in AML.


Sujet(s)
Régulation de l'expression des gènes dans la leucémie , Leucémie aigüe myéloïde/génétique , Protéines de maintenance des minichromosomes/génétique , Protéines de tissu nerveux/génétique , Facteurs d'épissage des ARN/génétique , Adénosine/analogues et dérivés , Adénosine/génétique , Animaux , Carcinogenèse/génétique , Lignée cellulaire tumorale , Réplication de l'ADN , Humains , Souris transgéniques , Composant-4 du complexe de maintenance des minichromosomes/génétique , Régulation positive
13.
Nucleic Acids Res ; 49(10): 5779-5797, 2021 06 04.
Article de Anglais | MEDLINE | ID: mdl-34048572

RÉSUMÉ

Faithful genome integrity maintenance plays an essential role in cell survival. Here, we identify the RNA demethylase ALKBH5 as a key regulator that protects cells from DNA damage and apoptosis during reactive oxygen species (ROS)-induced stress. We find that ROS significantly induces global mRNA N6-methyladenosine (m6A) levels by modulating ALKBH5 post-translational modifications (PTMs), leading to the rapid and efficient induction of thousands of genes involved in a variety of biological processes including DNA damage repair. Mechanistically, ROS promotes ALKBH5 SUMOylation through activating ERK/JNK signaling, leading to inhibition of ALKBH5 m6A demethylase activity by blocking substrate accessibility. Moreover, ERK/JNK/ALKBH5-PTMs/m6A axis is activated by ROS in hematopoietic stem/progenitor cells (HSPCs) in vivo in mice, suggesting a physiological role of this molecular pathway in the maintenance of genome stability in HSPCs. Together, our study uncovers a molecular mechanism involving ALKBH5 PTMs and increased mRNA m6A levels that protect genomic integrity of cells in response to ROS.


Sujet(s)
AlkB Homolog 5, RNA demethylase/métabolisme , Altération de l'ADN , Réparation de l'ADN , Espèces réactives de l'oxygène/métabolisme , AlkB Homolog 5, RNA demethylase/génétique , Animaux , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/métabolisme , Lignée cellulaire tumorale , Altération de l'ADN/effets des médicaments et des substances chimiques , Réparation de l'ADN/effets des médicaments et des substances chimiques , Réparation de l'ADN/génétique , Déméthylation/effets des médicaments et des substances chimiques , Protéines des groupes de complémentation de l'anémie de Fanconi/génétique , Protéines des groupes de complémentation de l'anémie de Fanconi/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/génétique , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/métabolisme , Humains , Peroxyde d'hydrogène/pharmacologie , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Méthylation/effets des médicaments et des substances chimiques , Souris , Phosphorylation , Maturation post-traductionnelle des protéines , Petit ARN interférent , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , RNA-Seq , Sumoylation/effets des médicaments et des substances chimiques , Spectrométrie de masse en tandem , Protéine nucléaire liée à l'X/génétique , Protéine nucléaire liée à l'X/métabolisme
14.
Blood ; 137(5): 610-623, 2021 02 04.
Article de Anglais | MEDLINE | ID: mdl-33538795

RÉSUMÉ

This study was conducted to determine the dosage effect of c-Myc on hematopoiesis and its distinct role in mediating the Wnt/ß-catenin pathway in hematopoietic stem cell (HSC) and bone marrow niche cells. c-Myc haploinsufficiency led to ineffective hematopoiesis by inhibiting HSC self-renewal and quiescence and by promoting apoptosis. We have identified Nr4a1, Nr4a2, and Jmjd3, which are critical for the maintenance of HSC functions, as previously unrecognized downstream targets of c-Myc in HSCs. c-Myc directly binds to the promoter regions of Nr4a1, Nr4a2, and Jmjd3 and regulates their expression. Our results revealed that Nr4a1 and Nr4a2 mediates the function of c-Myc in regulating HSC quiescence, whereas all 3 genes contribute to the function of c-Myc in the maintenance of HSC survival. Adenomatous polyposis coli (Apc) is a negative regulator of the Wnt/ß-catenin pathway. We have provided the first evidence that Apc haploinsufficiency induces a blockage of erythroid lineage differentiation through promoting secretion of IL6 in bone marrow endothelial cells. We found that c-Myc haploinsufficiency failed to rescue defective function of Apc-deficient HSCs in vivo but it was sufficient to prevent the development of severe anemia in Apc-heterozygous mice and to significantly prolong the survival of those mice. Furthermore, we showed that c-Myc-mediated Apc loss induced IL6 secretion in endothelial cells, and c-Myc haploinsufficiency reversed the negative effect of Apc-deficient endothelial cells on erythroid cell differentiation. Our studies indicate that c-Myc has a context-dependent role in mediating the function of Apc in hematopoiesis.


Sujet(s)
Gènes myc , Hématopoïèse/physiologie , Protéines proto-oncogènes c-myb/physiologie , Protéine de la polypose adénomateuse colique/physiologie , Anémie/génétique , Anémie/prévention et contrôle , Animaux , Apoptose/physiologie , Transplantation de moelle osseuse , Auto-renouvellement cellulaire/physiologie , Test clonogénique , Cellules endothéliales/anatomopathologie , Cellules érythroïdes/anatomopathologie , Délétion de gène , Gènes APC , Haploinsuffisance , Hématopoïèse/génétique , Cellules souches hématopoïétiques , Interleukine-6/physiologie , Jumonji Domain-Containing Histone Demethylases/physiologie , Souches mutantes de souris , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/physiologie , Membre-2 du groupe A de la sous-famille-4 de récepteurs nucléaires/physiologie , Poly I-C/pharmacologie , Chimère post-radique , Voie de signalisation Wnt/physiologie
15.
Front Physiol ; 11: 590180, 2020.
Article de Anglais | MEDLINE | ID: mdl-33101065

RÉSUMÉ

TFII-I is a ubiquitously expressed transcription factor that positively or negatively regulates gene expression. TFII-I has been implicated in neuronal and immunologic diseases as well as in thymic epithelial cancer. Williams-Beuren Syndrome (WBS) is caused by a large hemizygous deletion on chromosome 7q11.23 which encompasses 26-28 genes, including GTF2I, the human gene encoding TFII-I. A subset of WBS patients has recently been shown to present with macrocytosis, a mild anemia characterized by enlarged erythrocytes. We conditionally deleted the TFII-I/Gtf2i gene in adult mice by tamoxifen induced Cre-recombination. Bone marrow cells revealed defects in erythro-megakaryopoiesis and an increase in expression of the adult ß-globin gene. The data show that TFII-I acts as a repressor of ß-globin gene transcription and that it is implicated in the differentiation of erythro-megakaryocytic cells.

16.
Cell Stem Cell ; 27(1): 64-80.e9, 2020 07 02.
Article de Anglais | MEDLINE | ID: mdl-32402250

RÉSUMÉ

N6-methyladenosine (m6A), the most abundant internal modification in mRNA, has been implicated in tumorigenesis. As an m6A demethylase, ALKBH5 has been shown to promote the development of breast cancer and brain tumors. However, in acute myeloid leukemia (AML), ALKBH5 was reported to be frequently deleted, implying a tumor-suppressor role. Here, we show that ALKBH5 deletion is rare in human AML; instead, ALKBH5 is aberrantly overexpressed in AML. Moreover, its increased expression correlates with poor prognosis in AML patients. We demonstrate that ALKBH5 is required for the development and maintenance of AML and self-renewal of leukemia stem/initiating cells (LSCs/LICs) but not essential for normal hematopoiesis. Mechanistically, ALKBH5 exerts tumor-promoting effects in AML by post-transcriptional regulation of its critical targets such as TACC3, a prognosis-associated oncogene in various cancers. Collectively, our findings reveal crucial functions of ALKBH5 in leukemogenesis and LSC/LIC self-renewal/maintenance and highlight the therapeutic potential of targeting the ALKBH5/m6A axis.


Sujet(s)
Auto-renouvellement cellulaire , Leucémie aigüe myéloïde , AlkB Homolog 5, RNA demethylase/génétique , Carcinogenèse/génétique , Humains , Leucémie aigüe myéloïde/génétique , Protéines associées aux microtubules , Cellules souches tumorales
17.
Nat Commun ; 11(1): 928, 2020 02 17.
Article de Anglais | MEDLINE | ID: mdl-32066721

RÉSUMÉ

FOXM1, a known transcription factor, promotes cell proliferation in a variety of cancer cells. Here we show that Foxm1 is required for survival, quiescence and self-renewal of MLL-AF9 (MA9)-transformed leukemia stem cells (LSCs) in vivo. Mechanistically, Foxm1 upregulation activates the Wnt/ß-catenin signaling pathways by directly binding to ß-catenin and stabilizing ß-catenin protein through inhibiting its degradation, thereby preserving LSC quiescence, and promoting LSC self-renewal in MLL-rearranged AML. More importantly, inhibition of FOXM1 markedly suppresses leukemogenic potential and induces apoptosis of primary LSCs from MLL-rearranged AML patients in vitro and in vivo in xenograft mice. Thus, our study shows a critical role and mechanisms of Foxm1 in MA9-LSCs, and indicates that FOXM1 is a potential therapeutic target for selectively eliminating LSCs in MLL-rearranged AML.


Sujet(s)
Protéine M1 à motif en tête de fourche/métabolisme , Régulation de l'expression des gènes dans la leucémie , Leucémie aigüe myéloïde/génétique , Cellules souches tumorales/anatomopathologie , Animaux , Apoptose/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Protéine M1 à motif en tête de fourche/antagonistes et inhibiteurs , Protéine M1 à motif en tête de fourche/génétique , Réarrangement des gènes , Humains , Leucémie aigüe myéloïde/anatomopathologie , Souris , Souris knockout , Protéine de la leucémie myéloïde-lymphoïde/génétique , Protéines de fusion oncogènes/génétique , Culture de cellules primaires , RNA-Seq , Régulation positive , Voie de signalisation Wnt/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
18.
Leukemia ; 34(2): 380-390, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31492896

RÉSUMÉ

Leukemia stem cells (LSCs) are the rare populations of acute myeloid leukemia (AML) cells that are able to initiate, maintain, and propagate AML. Targeting LSCs is a promising approach for preventing AML relapse and improving long-term outcomes. While Slug, a zinc-finger transcription repressor, negatively regulates the self-renewal of normal hematopoietic stem cells, its functions in AML are still unknown. We report here that Slug promotes leukemogenesis and its loss impairs LSC self-renewal and delays leukemia progression. Mechanistically, Slc13a3, a direct target of Slug in LSCs, restricts the self-renewal of LSCs and markedly prolongs recipient survival. Genetic or pharmacological inhibition of SLUG or forced expression of Slc13a3 suppresses the growth of human AML cells. In conclusion, our studies demonstrate that Slug differentially regulates self-renewal of LSCs and normal HSCs, and both Slug and Slc13a3 are potential therapeutic targets of LSCs.


Sujet(s)
Leucémie aigüe myéloïde/métabolisme , Cellules souches tumorales/métabolisme , Espèces réactives de l'oxygène/métabolisme , Transduction du signal/physiologie , Facteurs de transcription de la famille Snail/métabolisme , Symporteurs/métabolisme , Animaux , Prolifération cellulaire/physiologie , Cellules souches hématopoïétiques/métabolisme , Humains , Souris , Souris de lignée C57BL , Tests d'activité antitumorale sur modèle de xénogreffe
19.
Cancer Cell ; 35(4): 677-691.e10, 2019 04 15.
Article de Anglais | MEDLINE | ID: mdl-30991027

RÉSUMÉ

FTO, an mRNA N6-methyladenosine (m6A) demethylase, was reported to promote leukemogenesis. Using structure-based rational design, we have developed two promising FTO inhibitors, namely FB23 and FB23-2, which directly bind to FTO and selectively inhibit FTO's m6A demethylase activity. Mimicking FTO depletion, FB23-2 dramatically suppresses proliferation and promotes the differentiation/apoptosis of human acute myeloid leukemia (AML) cell line cells and primary blast AML cells in vitro. Moreover, FB23-2 significantly inhibits the progression of human AML cell lines and primary cells in xeno-transplanted mice. Collectively, our data suggest that FTO is a druggable target and that targeting FTO by small-molecule inhibitors holds potential to treat AML.


Sujet(s)
Alpha-ketoglutarate-dependent dioxygenase FTO/antagonistes et inhibiteurs , Antinéoplasiques/pharmacologie , Antienzymes/pharmacologie , Leucémie aigüe myéloïde/traitement médicamenteux , Alpha-ketoglutarate-dependent dioxygenase FTO/composition chimique , Alpha-ketoglutarate-dependent dioxygenase FTO/génétique , Alpha-ketoglutarate-dependent dioxygenase FTO/métabolisme , Animaux , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacocinétique , Apoptose/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Antienzymes/composition chimique , Antienzymes/pharmacocinétique , Humains , Leucémie aigüe myéloïde/enzymologie , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Méthylation , Souris de lignée BALB C , Souris de lignée NOD , Souris transgéniques , Thérapie moléculaire ciblée , Conformation des protéines , ARN messager/génétique , ARN messager/métabolisme , Rat Sprague-Dawley , Transduction du signal , Relation structure-activité , Cellules U937 , Tests d'activité antitumorale sur modèle de xénogreffe
20.
Mol Cancer ; 18(1): 30, 2019 02 27.
Article de Anglais | MEDLINE | ID: mdl-30813931

RÉSUMÉ

Ectopic Viral Integration site 1 (EVI1) upregulation is implicated in 10-25% of pediatric acute myeloid leukemia (AML) and has an inferior outcome with current chemotherapy regimens. Here we report that EVI1 upregulation is associated with methylation of the miR-9 promoter and correlated with downregulation of miR-9 in human AML cell lines and bone marrow (BM) cells from pediatric patients. Reactivation of miR-9 by hypomethylating agents and forced expression of miR-9 in EVI1high leukemia cell lines and primary leukemia cells results in apoptosis and decreased proliferation of EVI1high leukemia cells. Furthermore, re-expression of miR-9 delays disease progression in EVI1high leukemia-xenograft mice. Our results suggest that EVI1-induced hypermethylation and downregulation of the miR-9 plays an important role in leukemogenesis in EVI-1high pediatric AML, indicating that hypomethylating agents may be a potential therapeutic strategy for EVI1high pediatric AML.


Sujet(s)
Épigenèse génétique , Régulation de l'expression des gènes dans la leucémie , Leucémie aigüe myéloïde/génétique , Protéine du locus du complexe MDS1 et EVI1/génétique , microARN/génétique , Animaux , Antimétabolites antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Lignée cellulaire tumorale , Prolifération cellulaire , Enfant , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Décitabine/pharmacologie , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/mortalité , Leucémie aigüe myéloïde/anatomopathologie , Protéine du locus du complexe MDS1 et EVI1/métabolisme , Souris , microARN/agonistes , microARN/antagonistes et inhibiteurs , microARN/métabolisme , Oligoribonucléotides/génétique , Oligoribonucléotides/métabolisme , Transduction du signal , Analyse de survie , Tests d'activité antitumorale sur modèle de xénogreffe
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...