Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 232
Filtrar
1.
Cell Rep Med ; 4(10): 101214, 2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37794587

RESUMO

Multiple myeloma (MM) growth is supported by an immune-tolerant bone marrow microenvironment. Here, we find that loss of Never in mitosis gene A (NIMA)-related kinase 2 (NEK2) in tumor microenvironmental cells is associated with MM growth suppression. The absence of NEK2 leads to both fewer tumor-associated macrophages (TAMs) and inhibitory T cells. NEK2 expression in myeloid progenitor cells promotes the generation of functional TAMs when stimulated with MM conditional medium. Clinically, high NEK2 expression in MM cells is associated with increased CD8+ T effector memory cells, while low NEK2 is associated with an IFN-γ gene signature and activated T cell response. Inhibition of NEK2 upregulates PD-L1 expression in MM cells and myeloid cells. In a mouse model, the combination of NEK2 inhibitor INH154 with PD-L1 blockade effectively eliminates MM cells and prolongs survival. Our results provide strong evidence that NEK2 inhibition may overcome tumor immune escape and support its further clinical development.


Assuntos
Mieloma Múltiplo , Camundongos , Animais , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Antígeno B7-H1/genética , Linfócitos T/metabolismo , Linhagem Celular Tumoral , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patologia , Microambiente Tumoral
3.
Sci Rep ; 11(1): 24345, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34934133

RESUMO

Mutations in the NF1 tumor suppressor gene are linked to arteriopathy. Nf1 heterozygosity (Nf1+/-) results in robust neointima formation, similar to humans, and myeloid-restricted Nf1+/- recapitulates this phenotype via MEK-ERK activation. Here we define the contribution of myeloid subpopulations to NF1 arteriopathy. Neutrophils from WT and Nf1+/- mice were functionally assessed in the presence of MEK and farnesylation inhibitors in vitro and neutrophil recruitment to lipopolysaccharide was assessed in WT and Nf1+/- mice. Littermate 12-15 week-old male wildtype and Nf1+/- mice were subjected to carotid artery ligation and provided either a neutrophil depleting antibody (1A8), liposomal clodronate to deplete monocytes/macrophages, or PD0325901 and neointima size was assessed 28 days after injury. Bone marrow transplant experiments assessed monocyte/macrophage mobilization during neointima formation. Nf1+/- neutrophils exhibit enhanced proliferation, migration, and adhesion via p21Ras activation of MEK in vitro and in vivo. Neutrophil depletion suppresses circulating Ly6Clow monocytes and enhances neointima size, while monocyte/macrophage depletion and deletion of CCR2 in bone marrow cells abolish neointima formation in Nf1+/- mice. Taken together, these findings suggest that neurofibromin-MEK-ERK activation in circulating neutrophils and monocytes during arterial remodeling is nuanced and points to important cross-talk between these populations in the pathogenesis of NF1 arteriopathy.


Assuntos
Lesões das Artérias Carótidas/patologia , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Células Progenitoras Mieloides/patologia , Neointima/patologia , Neurofibromatose 1/patologia , Neurofibromina 1/fisiologia , Receptores CCR2/fisiologia , Animais , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Lesões das Artérias Carótidas/etiologia , Lesões das Artérias Carótidas/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Monócitos/patologia , Células Progenitoras Mieloides/metabolismo , Neointima/etiologia , Neointima/metabolismo , Neurofibromatose 1/etiologia , Neurofibromatose 1/metabolismo
4.
Nat Commun ; 12(1): 7019, 2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34857757

RESUMO

Yolk sac (YS) hematopoiesis is critical for the survival of the embryo and a major source of tissue-resident macrophages that persist into adulthood. Yet, the transcriptional and epigenetic regulation of YS hematopoiesis remains poorly characterized. Here we report that the epigenetic regulator Ezh2 is essential for YS hematopoiesis but dispensable for subsequent aorta-gonad-mesonephros (AGM) blood development. Loss of EZH2 activity in hemogenic endothelium (HE) leads to the generation of phenotypically intact but functionally deficient erythro-myeloid progenitors (EMPs), while the generation of primitive erythroid cells is not affected. EZH2 activity is critical for the generation of functional EMPs at the onset of the endothelial-to-hematopoietic transition but subsequently dispensable. We identify a lack of Wnt signaling downregulation as the primary reason for the production of non-functional EMPs. Together, our findings demonstrate a critical and stage-specific role of Ezh2 in modulating Wnt signaling during the generation of EMPs from YS HE.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/genética , Células Eritroides/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Embrionárias Murinas/metabolismo , Células Progenitoras Mieloides/metabolismo , Proteínas de Transporte Vesicular/genética , Saco Vitelino/metabolismo , Animais , Diferenciação Celular , Embrião de Mamíferos , Proteína Potenciadora do Homólogo 2 de Zeste/deficiência , Epigênese Genética , Células Eritroides/citologia , Feminino , Feto , Genes Reporter , Hematopoese/genética , Fígado/citologia , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Embrionárias Murinas/citologia , Células Progenitoras Mieloides/patologia , Cultura Primária de Células , Proteínas de Transporte Vesicular/metabolismo , Via de Sinalização Wnt , Saco Vitelino/citologia , Saco Vitelino/crescimento & desenvolvimento , Proteína Vermelha Fluorescente
5.
Nat Commun ; 12(1): 6154, 2021 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-34686664

RESUMO

Acute myeloid leukemia (AML) harboring inv(16)(p13q22) expresses high levels of miR-126. Here we show that the CBFB-MYH11 (CM) fusion gene upregulates miR-126 expression through aberrant miR-126 transcription and perturbed miR-126 biogenesis via the HDAC8/RAN-XPO5-RCC1 axis. Aberrant miR-126 upregulation promotes survival of leukemia-initiating progenitors and is critical for initiating and maintaining CM-driven AML. We show that miR-126 enhances MYC activity through the SPRED1/PLK2-ERK-MYC axis. Notably, genetic deletion of miR-126 significantly reduces AML rate and extends survival in CM knock-in mice. Therapeutic depletion of miR-126 with an anti-miR-126 (miRisten) inhibits AML cell survival, reduces leukemia burden and leukemia stem cell (LSC) activity in inv(16) AML murine and xenograft models. The combination of miRisten with chemotherapy further enhances the anti-leukemia and anti-LSC activity. Overall, this study provides molecular insights for the mechanism and impact of miR-126 dysregulation in leukemogenesis and highlights the potential of miR-126 depletion as a therapeutic approach for inv(16) AML.


Assuntos
Antineoplásicos/uso terapêutico , Cromossomos Humanos Par 16/genética , Leucemia Mieloide Aguda/tratamento farmacológico , MicroRNAs/antagonistas & inibidores , Células-Tronco Neoplásicas/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ciclo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Inversão Cromossômica/genética , Família de Proteínas EGF/genética , Fator de Transcrição GATA2/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Histona Desacetilases/metabolismo , Humanos , Carioferinas/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Terapia de Alvo Molecular , Células Progenitoras Mieloides/efeitos dos fármacos , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Proteínas Nucleares/metabolismo , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Proteínas Repressoras/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína ran de Ligação ao GTP/metabolismo
7.
Elife ; 102021 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-33830019

RESUMO

Innate immune cellular effectors are actively consumed during systemic inflammation, but the systemic traffic and the mechanisms that support their replenishment remain unknown. Here, we demonstrate that acute systemic inflammation induces the emergent activation of a previously unrecognized system of rapid migration of granulocyte-macrophage progenitors and committed macrophage-dendritic progenitors, but not other progenitors or stem cells, from bone marrow (BM) to regional lymphatic capillaries. The progenitor traffic to the systemic lymphatic circulation is mediated by Ccl19/Ccr7 and is NF-κB independent, Traf6/IκB-kinase/SNAP23 activation dependent, and is responsible for the secretion of pre-stored Ccl19 by a subpopulation of CD205+/CD172a+ conventional dendritic cells type 2 and upregulation of BM myeloid progenitor Ccr7 signaling. Mature myeloid Traf6 signaling is anti-inflammatory and necessary for lymph node myeloid cell development. This report unveils the existence and the mechanistic basis of a very early direct traffic of myeloid progenitors from BM to lymphatics during inflammation.


When the body becomes infected with disease-causing pathogens, such as bacteria, the immune system activates various mechanisms which help to fight off the infection. One of the immune system's first lines of defense is to launch an inflammatory response that helps remove the pathogen and recruit other immune cells. However, this response can become overactivated, leading to severe inflammatory conditions that damage healthy cells and tissues. A second group of cells counteract this over inflammation and are different to the ones involved in the early inflammatory response. Both types of cells ­ inflammatory and anti-inflammatory ­ develop from committed progenitors, which, unlike stem cells, are already destined to become a certain type of cell. These committed progenitors reside in the bone marrow and then rapidly travel to secondary lymphoid organs, such as the lymph nodes, where they mature into functioning immune cells. During this journey, committed progenitors pass from the bone marrow to the lymphatic vessels that connect up the different secondary lymphoid organs, and then spread to all tissues in the body. Yet, it is not fully understood what exact route these cells take and what guides them towards these lymphatic tissues during inflammation. To investigate this, Serrano-Lopez, Hegde et al. used a combination of techniques to examine the migration of progenitor cells in mice that had been treated with lethal doses of a bacterial product that triggers inflammation. This revealed that as early as one to three hours after the onset of infection, progenitor cells were already starting to travel from the bone marrow towards lymphatic vessels. Serrano-Lopez, Hegde et al. found that a chemical released by an "alarm" immune cell already residing in secondary lymphoid organs attracted these progenitor cells towards the lymphatic tissue. Further experiments showed that the progenitor cells travelling to secondary lymphoid organs were already activated by bacterial products. They then follow the chemical released by alarm immune cells ready to respond to the immune challenge and suppress inflammation. These committed progenitors were also found in the inflamed lymph nodes of patients. These findings suggest this rapid circulation of progenitors is a mechanism of defense that contributes to the fight against severe inflammation. Altering how these cells migrate from the bone marrow to secondary lymphoid organs could provide a more effective treatment for inflammatory conditions and severe infections. However, these approaches would need to be tested further in the laboratory and in clinical trials.


Assuntos
Medula Óssea/metabolismo , Movimento Celular , Células Progenitoras de Granulócitos e Macrófagos/metabolismo , Mediadores da Inflamação/metabolismo , Inflamação/metabolismo , Linfadenopatia/metabolismo , Sistema Linfático/metabolismo , Células Progenitoras Mieloides/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Medula Óssea/imunologia , Medula Óssea/patologia , Linhagem da Célula , Células Cultivadas , Criança , Pré-Escolar , Modelos Animais de Doenças , Feminino , Células Progenitoras de Granulócitos e Macrófagos/imunologia , Células Progenitoras de Granulócitos e Macrófagos/patologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Linfadenopatia/imunologia , Linfadenopatia/patologia , Sistema Linfático/imunologia , Sistema Linfático/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Células Progenitoras Mieloides/imunologia , Células Progenitoras Mieloides/patologia , Fenótipo , Transdução de Sinais , Fatores de Tempo , Adulto Jovem
8.
Nat Chem Biol ; 17(5): 567-575, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33664520

RESUMO

The discovery of effective therapeutic treatments for cancer via cell differentiation instead of antiproliferation remains a great challenge. Cyclin-dependent kinase 2 (CDK2) inactivation, which overcomes the differentiation arrest of acute myeloid leukemia (AML) cells, may be a promising method for AML treatment. However, there is no available selective CDK2 inhibitor. More importantly, the inhibition of only the enzymatic function of CDK2 would be insufficient to promote notable AML differentiation. To further validate the role and druggability of CDK2 involved in AML differentiation, a suitable chemical tool is needed. Therefore, we developed first-in-class CDK2-targeted proteolysis-targeting chimeras (PROTACs), which promoted rapid and potent CDK2 degradation in different cell lines without comparable degradation of other targets, and induced remarkable differentiation of AML cell lines and primary patient cells. These data clearly demonstrated the practicality and importance of PROTACs as alternative tools for verifying CDK2 protein functions.


Assuntos
Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Células Progenitoras Mieloides/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Triazóis/farmacologia , Antineoplásicos/síntese química , Aurora Quinase A/genética , Aurora Quinase A/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/metabolismo , Desenho de Fármacos , Descoberta de Drogas , Humanos , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/metabolismo , Concentração Inibidora 50 , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/enzimologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Células Progenitoras Mieloides/enzimologia , Células Progenitoras Mieloides/patologia , Piperazinas/farmacologia , Cultura Primária de Células , Piridinas/farmacologia , Pirimidinas/farmacologia , Quinazolinas/farmacologia , Transdução de Sinais , Relação Estrutura-Atividade , Transcriptoma , Triazóis/síntese química
10.
Br J Haematol ; 192(6): 1054-1063, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33618432

RESUMO

Clonal haematopoiesis (CH) in patients with acute myeloid leukaemia (AML) may persist beyond attaining complete remission. From a consecutive cohort of 67 patients with nucleophosmin 1-mutated (NPM1mut ) AML, we identified 50 who achieved NPM1mut clearance and had parallel multicolour flow cytometry (MFC) and next generation sequencing (NGS). In total, 13 (26%) cleared all mutations, 37 (74%) had persistent CH frequently involving DNA methyltransferase 3α (DNMT3A,70%), tet methylcytosine dioxygenase 2 (TET2, 27%), isocitrate dehydrogenase 2 (IDH2, 19%) and IDH1 (11%). A small number (<1%) of aberrant CD34+ myeloblasts, but immunophenotypically different from original AML blasts [herein referred to as a pre-leukaemic (PL) phenotype], was detected in 17 (49%) patients with CH, but not in any patients with complete clearance of all mutations (P = 0·0037). A PL phenotype was associated with higher mutation burden (P = 0·005). Persistent IDH2 and serine and arginine-rich splicing factor 2 (SRSF2) mutations were exclusively observed in PL+ CH+ cases (P = 0·016). Persistent dysplasia was seen exclusively in cases with a PL+ phenotype (29% vs. none; P = 0·04). The PL+ phenotype did not correlate with age, intensity of induction therapy or relapse-free survival. Post-remission CH in the setting of NPM1mut clearance is common and may result in immunophenotypic changes in myeloid progenitors. It is important to not misinterpret these cells as AML measurable residual disease (MRD).


Assuntos
Medula Óssea , Hematopoiese Clonal , Leucemia Mieloide Aguda , Mutação , Células Progenitoras Mieloides , Proteínas de Neoplasias , Proteínas Nucleares , Adulto , Idoso , Idoso de 80 Anos ou mais , Medula Óssea/metabolismo , Medula Óssea/patologia , Feminino , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Leucemia Mieloide Aguda/terapia , Masculino , Pessoa de Meia-Idade , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Indução de Remissão
11.
Sci Rep ; 10(1): 21804, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33311522

RESUMO

Previous studies examining the role of the histone deacetylase Hdac3 within myeloid cells demonstrated that Hdac3 promotes M2 activation and tissue healing in inflammatory conditions. Since myeloid lineage cells are required for proper bone formation and regeneration, in this study we examined the functions of Hdac3 during bone healing. Conditional deletion of Hdac3 within myeloid progenitors accelerates healing of cortical bone defects. Moreover, reduced osteoclast numbers within the defect site are correlated with Hdac3 suppression. Ex vivo osteoclastogenesis assays further demonstrate that Hdac3 deficiency limits osteoclastogenesis, the number of nuclei per cell and bone resorption, suggesting a defect in cell fusion. High throughput RNA sequencing identified the transmembrane protein Pmepa1 as a differentially expressed gene within osteoclast progenitor cells. Knockdown of Pmepa1 partially restores defects in osteoclastogenesis induced by Hdac3 deficiency. These results show that Hdac3 is required for optimal bone healing and osteoclast fusion, potentially via its regulation of Pmepa1 expression.


Assuntos
Regeneração Óssea , Osso Cortical/metabolismo , Deleção de Genes , Histona Desacetilases/deficiência , Proteínas de Membrana/metabolismo , Células Progenitoras Mieloides/metabolismo , Osteoclastos/metabolismo , Animais , Fusão Celular , Osso Cortical/lesões , Osso Cortical/patologia , Feminino , Histona Desacetilases/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Células Progenitoras Mieloides/patologia , Osteoclastos/patologia
12.
BMC Genomics ; 21(1): 652, 2020 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-32967610

RESUMO

BACKGROUND: The identification of cell type-specific genes (markers) is an essential step for the deconvolution of the cellular fractions, primarily, from the gene expression data of a bulk sample. However, the genes with significant changes identified by pair-wise comparisons cannot indeed represent the specificity of gene expression across multiple conditions. In addition, the knowledge about the identification of gene expression markers across multiple conditions is still paucity. RESULTS: Herein, we developed a hybrid tool, LinDeconSeq, which consists of 1) identifying marker genes using specificity scoring and mutual linearity strategies across any number of cell types, and 2) predicting cellular fractions of bulk samples using weighted robust linear regression with the marker genes identified in the first stage. On multiple publicly available datasets, the marker genes identified by LinDeconSeq demonstrated better accuracy and reproducibility compared to MGFM and RNentropy. Among deconvolution methods, LinDeconSeq showed low average deviations (≤0.0958) and high average Pearson correlations (≥0.8792) between the predicted and actual fractions on the benchmark datasets. Importantly, the cellular fractions predicted by LinDeconSeq appear to be relevant in the diagnosis of acute myeloid leukemia (AML). The distinct cellular fractions in granulocyte-monocyte progenitor (GMP), lymphoid-primed multipotent progenitor (LMPP) and monocytes (MONO) were found to be closely associated with AML compared to the healthy samples. Moreover, the heterogeneity of cellular fractions in AML patients divided these patients into two subgroups, differing in both prognosis and mutation patterns. GMP fraction was the most pronounced between these two subgroups, particularly, in SubgroupA, which was strongly associated with the better AML prognosis and the younger population. Totally, the identification of marker genes by LinDeconSeq represents the improved feature for deconvolution. The data processing strategy with regard to the cellular fractions used in this study also showed potential for the diagnosis and prognosis of diseases. CONCLUSIONS: Taken together, we developed a freely-available and open-source tool LinDeconSeq ( https://github.com/lihuamei/LinDeconSeq ), which includes marker identification and deconvolution procedures. LinDeconSeq is comparable to other current methods in terms of accuracy when applied to benchmark datasets and has broad application in clinical outcome and disease-specific molecular mechanisms.


Assuntos
Biomarcadores Tumorais/genética , Leucemia Mieloide Aguda/genética , Células Progenitoras Mieloides/classificação , Software , Humanos , Leucemia Mieloide Aguda/classificação , Leucemia Mieloide Aguda/patologia , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patologia
13.
Blood ; 136(19): 2188-2199, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-32750121

RESUMO

Langerhans cell histiocytosis (LCH) is a myeloid neoplasia, driven by sporadic activating mutations in the MAPK pathway. The misguided myeloid dendritic cell (DC) model proposes that high-risk, multisystem, risk-organ-positive (MS-RO+) LCH results from driver mutation in a bone marrow (BM)-resident multipotent hematopoietic progenitor, while low-risk, MS-RO- and single-system LCH would result from driver mutation in a circulating or tissue-resident, DC-committed precursor. We have examined the CD34+c-Kit+Flt3+ myeloid progenitor population as potential mutation carrier in all LCH disease manifestations. This population contains oligopotent progenitors of monocytes (Mo's)/macrophages (MΦs), osteoclasts (OCs), and DCs. CD34+c-Kit+Flt3+ cells from BM of MS-RO+ LCH patients produced Langerhans cell (LC)-like cells in vitro. Both LC-like and DC offspring from this progenitor carried the BRAF mutation, confirming their common origin. In both high- and low-risk LCH patients, CD34+c-Kit+Flt3+ progenitor frequency in blood was higher than in healthy donors. In one MS-RO+ LCH patient, CD34+c-Kit+Flt3+ cell frequency in blood and its BRAF-mutated offspring reported response to chemotherapy. CD34+c-Kit+Flt3+ progenitors from blood of both high- and low-risk LCH patients gave rise to DCs and LC-like cells in vitro, but the driver mutation was not easily detectable, likely due to low frequency of mutated progenitors. Mutant BRAF alleles were found in Mo's /MΦs, DCs, LC-like cells, and/or OC-like cells in lesions and/or Mo and DCs in blood of multiple low-risk patients. We therefore hypothesize that in both high- and low-risk LCH, the driver mutation is present in a BM-resident myeloid progenitor that can be mobilized to the blood.


Assuntos
Medula Óssea/patologia , Diferenciação Celular , Células Dendríticas/patologia , Histiocitose de Células de Langerhans/patologia , Mutação , Células Progenitoras Mieloides/patologia , Proteínas Proto-Oncogênicas B-raf/genética , Medula Óssea/metabolismo , Células Dendríticas/metabolismo , Histiocitose de Células de Langerhans/genética , Histiocitose de Células de Langerhans/metabolismo , Humanos , Células Progenitoras Mieloides/metabolismo
14.
Stem Cell Reports ; 15(1): 6-12, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32559456

RESUMO

Specific subgroups of acute myeloid leukemia (AML), including those containing MLL rearrangements and NPM1c mutations, possess characteristic stem cell-like gene expression profiles. These expression programs are highly dependent on components of the MLL histone methyltransferase complex, including Menin and DOT1L. Understanding the chromatin-based mechanisms through which cancer cells subvert certain aspects of normal stem cell biology helped identify specific vulnerabilities and translate them into targeted therapy approaches. Exciting progress has been made in the development of small-molecule inhibitors targeting this epigenetic machinery in leukemia cells and prompted the development of clinical trials in patients with hematologic malignancies.


Assuntos
Autorrenovação Celular , Cromatina/metabolismo , Leucemia Mieloide Aguda/patologia , Células Progenitoras Mieloides/patologia , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Terapia de Alvo Molecular , Células-Tronco Neoplásicas/patologia
15.
Blood ; 136(2): 183-198, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32305041

RESUMO

Hematopoietic stem cell (HSC) aging correlates with an increasing risk of myeloproliferative disease and immunosenescence. In this study, we show that aging-related inflammation promotes HSC aging through tumor necrosis factor-α (TNF-α)→ERK→ETS1→interleukin27Ra (IL27Ra) pathway. TNF-α, a well-known biomarker of inflammation, increases during aging and induces the expression of IL27Ra on HSCs via ERK-ETS1 signaling. Deletion of IL27Ra rescues the functional decline and myeloid bias of HSCs and also reverses the inhibitory effect of TNF-α on HSCs. Aged IL27Ra-/- mice had a reduced proportion of myeloid-biased HSCs and did not display the biased myeloid differentiation that occurs in aged wild-type mice. IL27Ra+ HSCs exhibit impaired reconstitution capacity and myeloid-bias compared with IL27Ra- HSCs and serve as a myeloid-recovery pool upon inflammatory insult. Inflammation-related genes were enriched in IL27Ra+ HSCs and this enrichment increases with aging. Our study demonstrates that age-induced IL27Ra signaling impairs HSCs and raises the possibility that interfering with IL27Ra signaling can counter the physiologically deleterious effect of aging on hematopoietic capacity.


Assuntos
Envelhecimento/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Células Progenitoras Mieloides/imunologia , Receptores de Interleucina/imunologia , Envelhecimento/genética , Envelhecimento/patologia , Animais , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Knockout , Células Progenitoras Mieloides/patologia , Receptores de Interleucina/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
16.
Oncogene ; 39(15): 3195-3205, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32115572

RESUMO

ZBTB7A is frequently mutated in acute myeloid leukemia (AML) with t(8;21) translocation. However, the oncogenic collaboration between mutated ZBTB7A and the RUNX1-RUNX1T1 fusion gene in AML t(8;21) remains unclear. Here, we investigate the role of ZBTB7A and its mutations in the context of normal and malignant hematopoiesis. We demonstrate that clinically relevant ZBTB7A mutations in AML t(8;21) lead to loss of function and result in perturbed myeloid differentiation with block of the granulocytic lineage in favor of monocytic commitment. In addition, loss of ZBTB7A increases glycolysis and hence sensitizes leukemic blasts to metabolic inhibition with 2-deoxy-D-glucose. We observed that ectopic expression of wild-type ZBTB7A prevents RUNX1-RUNX1T1-mediated clonal expansion of human CD34+ cells, whereas the outgrowth of progenitors is enabled by ZBTB7A mutation. Finally, ZBTB7A expression in t(8;21) cells lead to a cell cycle arrest that could be mimicked by inhibition of glycolysis. Our findings suggest that loss of ZBTB7A may facilitate the onset of AML t(8;21), and that RUNX1-RUNX1T1-rearranged leukemia might be treated with glycolytic inhibitors.


Assuntos
Carcinogênese/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Proteínas de Ligação a DNA/genética , Hematopoese/genética , Leucemia Mieloide Aguda/genética , Proteínas de Fusão Oncogênica/metabolismo , Proteína 1 Parceira de Translocação de RUNX1/metabolismo , Fatores de Transcrição/genética , Animais , Medula Óssea/patologia , Carcinogênese/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular Tumoral , Linhagem da Célula/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Proteínas de Ligação a DNA/metabolismo , Desoxiglucose/farmacologia , Desoxiglucose/uso terapêutico , Técnicas de Inativação de Genes , Glicólise/efeitos dos fármacos , Glicólise/genética , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/patologia , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Mutação com Perda de Função , Camundongos , Células Progenitoras Mieloides/patologia , Proteínas de Fusão Oncogênica/genética , Proteína 1 Parceira de Translocação de RUNX1/genética , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Transcrição/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Science ; 367(6477): 586-590, 2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-32001657

RESUMO

The initiating mutations that contribute to cancer development are sometimes present in premalignant cells. Whether therapies targeting these mutations can eradicate premalignant cells is unclear. Acute myeloid leukemia (AML) is an attractive system for investigating the effect of preventative treatment because this disease is often preceded by a premalignant state (clonal hematopoiesis or myelodysplastic syndrome). In Npm1c/Dnmt3a mutant knock-in mice, a model of AML development, leukemia is preceded by a period of extended myeloid progenitor cell proliferation and self-renewal. We found that this self-renewal can be reversed by oral administration of a small molecule (VTP-50469) that targets the MLL1-Menin chromatin complex. These preclinical results support the hypothesis that individuals at high risk of developing AML might benefit from targeted epigenetic therapy in a preventative setting.


Assuntos
Terapia Genética/métodos , Leucemia Experimental/prevenção & controle , Leucemia Mieloide Aguda/prevenção & controle , Proteínas Nucleares/genética , Pré-Leucemia/terapia , Animais , Cromatina/metabolismo , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Técnicas de Introdução de Genes , Histona-Lisina N-Metiltransferase/metabolismo , Leucemia Experimental/genética , Leucemia Mieloide Aguda/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação , Células Progenitoras Mieloides/patologia , Proteína de Leucina Linfoide-Mieloide/metabolismo , Nucleofosmina , Pré-Leucemia/genética , Pré-Leucemia/patologia , Proteínas Proto-Oncogênicas/metabolismo
18.
Exp Hematol ; 82: 43-52.e4, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32014431

RESUMO

Aged hematopoietic stem cells (HSCs) undergo biased lineage priming and differentiation toward production of myeloid cells. A comprehensive understanding of gene regulatory mechanisms causing HSC aging is needed to devise new strategies to sustainably improve immune function in aged individuals. Here, a focused short hairpin RNA screen of epigenetic factors reveals that the histone acetyltransferase Kat6b regulates myeloid cell production from hematopoietic progenitor cells. Within the stem and progenitor cell compartment, Kat6b is highly expressed in long-term (LT)-HSCs and is significantly decreased with aging at the transcript and protein levels. Knockdown of Kat6b in young LT-HSCs causes skewed production of myeloid cells at the expense of erythroid cells both in vitro and in vivo. Transcriptome analysis identifies enrichment of aging and macrophage-associated gene signatures alongside reduced expression of self-renewal and multilineage priming signatures. Together, our work identifies KAT6B as a novel epigenetic regulator of hematopoietic differentiation and a target to improve aged immune function.


Assuntos
Envelhecimento/metabolismo , Diferenciação Celular , Células Eritroides/enzimologia , Regulação Enzimológica da Expressão Gênica , Histona Acetiltransferases/biossíntese , Células Progenitoras Mieloides/enzimologia , Envelhecimento/genética , Envelhecimento/patologia , Animais , Epigênese Genética , Células Eritroides/patologia , Perfilação da Expressão Gênica , Técnicas de Inativação de Genes , Histona Acetiltransferases/genética , Masculino , Camundongos , Camundongos Transgênicos , Células Progenitoras Mieloides/patologia , Transcriptoma
19.
Blood ; 135(3): 167-180, 2020 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-31805184

RESUMO

NF-κB is a key regulator of inflammation and cancer progression, with an important role in leukemogenesis. Despite its therapeutic potential, targeting NF-κB using pharmacologic inhibitors has proven challenging. Here, we describe a myeloid cell-selective NF-κB inhibitor using an miR-146a mimic oligonucleotide conjugated to a scavenger receptor/Toll-like receptor 9 agonist (C-miR146a). Unlike an unconjugated miR146a, C-miR146a was rapidly internalized and delivered to the cytoplasm of target myeloid cells and leukemic cells. C-miR146a reduced expression of classic miR-146a targets (IRAK1 and TRAF6), thereby blocking activation of NF-κB in target cells. IV injections of C-miR146a mimic to miR-146a-deficient mice prevented excessive NF-κB activation in myeloid cells, and thus alleviated myeloproliferation and mice hypersensitivity to bacterial challenge. Importantly, C-miR146a showed efficacy in dampening severe inflammation in clinically relevant models of chimeric antigen receptor (CAR) T-cell-induced cytokine release syndrome. Systemic administration of C-miR146a oligonucleotide alleviated human monocyte-dependent release of IL-1 and IL-6 in a xenotransplanted B-cell lymphoma model without affecting CD19-specific CAR T-cell antitumor activity. Beyond anti-inflammatory functions, miR-146a is a known tumor suppressor commonly deleted or expressed at reduced levels in human myeloid leukemia. Using The Cancer Genome Atlas acute myeloid leukemia data set, we found an inverse correlation of miR-146a levels with NF-κB-related genes and with patient survival. Correspondingly, C-miR146a induced cytotoxic effects in human MDSL, HL-60, and MV4-11 leukemia cells in vitro. The repeated IV administration of C-miR146a inhibited expression of NF-κB target genes and thereby thwarted progression of disseminated HL-60 leukemia. Our results show the potential of using myeloid cell-targeted miR-146a mimics for the treatment of inflammatory and myeloproliferative disorders.


Assuntos
Síndrome da Liberação de Citocina/prevenção & controle , Inflamação/prevenção & controle , Leucemia Mieloide Aguda/prevenção & controle , MicroRNAs/genética , Células Progenitoras Mieloides/patologia , NF-kappa B/metabolismo , Animais , Apoptose , Proliferação de Células , Síndrome da Liberação de Citocina/genética , Síndrome da Liberação de Citocina/patologia , Feminino , Regulação da Expressão Gênica , Humanos , Inflamação/genética , Inflamação/patologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Células Progenitoras Mieloides/metabolismo , NF-kappa B/genética , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Nat Commun ; 10(1): 5767, 2019 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-31852898

RESUMO

Cancer is a hyper-proliferative disease. Whether the proliferative state originates from the cell-of-origin or emerges later remains difficult to resolve. By tracking de novo transformation from normal hematopoietic progenitors expressing an acute myeloid leukemia (AML) oncogene MLL-AF9, we reveal that the cell cycle rate heterogeneity among granulocyte-macrophage progenitors (GMPs) determines their probability of transformation. A fast cell cycle intrinsic to these progenitors provide permissiveness for transformation, with the fastest cycling 3% GMPs acquiring malignancy with near certainty. Molecularly, we propose that MLL-AF9 preserves gene expression of the cellular states in which it is expressed. As such, when expressed in the naturally-existing, rapidly-cycling immature myeloid progenitors, this cell state becomes perpetuated, yielding malignancy. In humans, high CCND1 expression predicts worse prognosis for MLL fusion AMLs. Our work elucidates one of the earliest steps toward malignancy and suggests that modifying the cycling state of the cell-of-origin could be a preventative approach against malignancy.


Assuntos
Transformação Celular Neoplásica/genética , Regulação Leucêmica da Expressão Gênica , Leucemia Mieloide Aguda/genética , Células Progenitoras Mieloides/patologia , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas de Fusão Oncogênica/genética , Animais , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Transformação Celular Neoplásica/efeitos dos fármacos , Ciclina D1/metabolismo , Modelos Animais de Doenças , Feminino , Técnicas de Introdução de Genes , Humanos , Estimativa de Kaplan-Meier , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/mortalidade , Masculino , Camundongos Transgênicos , Piperazinas/administração & dosagem , Cultura Primária de Células , Prognóstico , Piridinas/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...