Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 183
Filtrar
1.
Int J Mol Sci ; 25(4)2024 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-38396817

RESUMEN

Acute myeloid leukemia (AML) is an aggressive malignancy characterized by rapid growth and uncontrolled proliferation of undifferentiated myeloid cells. Metabolic reprogramming is commonly observed in the bone marrow of AML patients, as leukemia cells require increased ATP supply to support disease progression. In this study, we examined the potential role of mesothelin as a metabolic modulator in myeloid cells in AML. Mesothelin is a well-known marker of solid tumors that promotes cancer cell proliferation and survival. We initially analyzed alterations in mesothelin expression in the myeloblast subpopulations, defined as SSC-Alow/CD45dim, obtained from the bone marrow of AML patients using flow cytometry. Our results showed overexpression of mesothelin in 34.8% of AML patients. Subsequently, metabolic changes in leukemia cells were evaluated by comparing the oxygen consumption rates (OCR) of bone marrow samples derived from adult AML patients. Notably, a higher OCR was observed in the mesothelin-positive compared to the mesothelin-low and non-expressing groups. Treatment with recombinant human mesothelin protein enhanced OCR and increased the mRNA expression of glycolytic enzymes and mitochondrial complex II in KG1α AML cells. Notably, siRNA targeting mesothelin in KG1α cells led to the reduction of glycolysis-related gene expression but had no effect on the mitochondrial complex gene. The collective results demonstrate that mesothelin induces metabolic changes in leukemia cells, facilitating the acquisition of a rapid supply of ATP for proliferation in AML. Therefore, the targeting of mesothelin presents a potentially promising approach to mitigating the progression of AML through the inhibition of glycolysis and mitochondrial respiration in myeloid cells.


Asunto(s)
Leucemia Mieloide Aguda , Mesotelina , Adulto , Humanos , Células Precursoras de Granulocitos/metabolismo , Succinato Deshidrogenasa/metabolismo , Línea Celular Tumoral , Leucemia Mieloide Aguda/genética , Proliferación Celular , Respiración , Glucólisis , Adenosina Trifosfato/metabolismo
2.
Leukemia ; 37(9): 1850-1859, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37532789

RESUMEN

The transcription factor CCAAT-enhancer binding factor alpha (C/ebpα) is a master controller of myeloid differentiation that is expressed as long (p42) and short (p30) isoform. Mutations within the CEBPA gene selectively deleting p42 are frequent in human acute myeloid leukemia. Here we investigated the individual genomics and transcriptomics of p42 and p30. Both proteins bound to identical sites across the genome. For most targets, they induced a highly similar transcriptional response with the exception of a few isoform specific genes. Amongst those we identified early growth response 1 (Egr1) and tribbles1 (Trib1) as key targets selectively induced by p42 that are also underrepresented in CEBPA-mutated AML. Egr1 executed a program of myeloid differentiation and growth arrest. Oppositely, Trib1 established a negative feedback loop through activation of Erk1/2 kinase thus placing differentiation under control of signaling. Unexpectedly, differentiation elicited either by removal of an oncogenic input or by G-CSF did not peruse C/ebpα as mediator but rather directly affected the cell cycle core by upregulation of p21/p27 inhibitors. This points to functions downstream of C/ebpα as intersection point where transforming and differentiation stimuli converge and this finding offers a new perspective for therapeutic intervention.


Asunto(s)
Células Precursoras de Granulocitos , Leucemia Mieloide Aguda , Humanos , Células Precursoras de Granulocitos/metabolismo , Leucemia Mieloide Aguda/genética , Diferenciación Celular , Isoformas de Proteínas/genética , Mutación , Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo
3.
Int J Mol Sci ; 23(19)2022 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-36232688

RESUMEN

Acute myeloid leukemia (AML)-the most frequent form of adult blood cancer-is characterized by heterogeneous mechanisms and disease progression. Developing an effective therapeutic strategy that targets metabolic homeostasis and energy production in immature leukemic cells (blasts) is essential for overcoming relapse and improving the prognosis of AML patients with different subtypes. With respect to metabolic regulation, fructose-1,6-bisphosphatase 1 (FBP1) is a gluconeogenic enzyme that is vital to carbohydrate metabolism, since gluconeogenesis is the central pathway for the production of important metabolites and energy necessary to maintain normal cellular activities. Beyond its catalytic activity, FBP1 inhibits aerobic glycolysis-known as the "Warburg effect"-in cancer cells. Importantly, while downregulation of FBP1 is associated with carcinogenesis in major human organs, restoration of FBP1 in cancer cells promotes apoptosis and prevents disease progression in solid tumors. Recently, our large-scale sequencing analyses revealed FBP1 as a novel inducible therapeutic target among 17,757 vitamin-D-responsive genes in MV4-11 or MOLM-14 blasts in vitro, both of which were derived from AML patients with FLT3 mutations. To investigate FBP1's anti-leukemic function in this study, we generated a new AML cell line through lentiviral overexpression of an FBP1 transgene in vitro (named FBP1-MV4-11). Results showed that FBP1-MV4-11 blasts are more prone to apoptosis than MV4-11 blasts. Mechanistically, FBP1-MV4-11 blasts have significantly increased gene and protein expression of P53, as confirmed by the P53 promoter assay in vitro. However, enhanced cell death and reduced proliferation of FBP1-MV4-11 blasts could be reversed by supplementation with post-glycolytic metabolites in vitro. Additionally, FBP1-MV4-11 blasts were found to have impaired mitochondrial homeostasis through reduced cytochrome c oxidase subunit 2 (COX2 or MT-CO2) and upregulated PTEN-induced kinase (PINK1) expressions. In summary, this is the first in vitro evidence that FBP1-altered carbohydrate metabolism and FBP1-activated P53 can initiate leukemic death by activating mitochondrial reprogramming in AML blasts, supporting the clinical potential of FBP1-based therapies for AML-like cancers.


Asunto(s)
Metabolismo de los Hidratos de Carbono , Células Precursoras de Granulocitos , Leucemia Mieloide Aguda , Mitocondrias , Proteína p53 Supresora de Tumor , Apoptosis , Metabolismo de los Hidratos de Carbono/efectos de los fármacos , Metabolismo de los Hidratos de Carbono/genética , Dióxido de Carbono/metabolismo , Línea Celular Tumoral , Ciclooxigenasa 2/metabolismo , Progresión de la Enfermedad , Complejo IV de Transporte de Electrones/metabolismo , Fructosa/farmacología , Fructosa-Bifosfatasa/genética , Fructosa-Bifosfatasa/metabolismo , Glucólisis , Células Precursoras de Granulocitos/metabolismo , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Quinasas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Vitamina D/farmacología , Vitaminas/farmacología , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo
4.
Shock ; 57(5): 749-758, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35583915

RESUMEN

BACKGROUND: Intratracheal (IT) lipopolysaccharide (LPS) causes severe acute lung injury (ALI) and systemic inflammation. CMT-3 has pleiotropic anti-inflammatory effects including matrix metalloproteinase (MMP) inhibition, attenuation of neutrophil (PMN) activation, and elastase release. CMT-3's poor water solubility limits its bioavailability when administered orally for treating ALI. We developed a nano-formulation of CMT-3 (nCMT-3) to test the hypothesis that the pleiotropic anti-inflammatory activities of IT nCMT-3 can attenuate LPS-induced ALI. METHODS: C57BL/6 mice were treated with aerosolized IT nCMT-3 or saline, then had IT LPS or saline administered 2 h later. Tissues were harvested at 24 h. The effects of LPS and nCMT-3 on ALI were assessed by lung histology, MMP level/activity (zymography), NLRP3 protein, and activated caspase-1 levels. Blood and bronchoalveolar lavage fluid (BALF) cell counts, PMN elastase, and soluble triggering receptor expressed on myelocytes-1 (sTREM-1) levels, TNF-α, IL-1ß, IL-6, IL-18, and BALF protein levels were also measured. RESULTS: LPS-induced ALI was characterized by histologic lung injury (PMN infiltration, alveolar thickening, edema, and consolidation) elevated proMMP-2, -9 levels and activity, increased NLRP-3 protein and activated caspase-1 levels in lung tissue. LPS-induced increases in plasma and BALF levels of sTREM-1, TNF-α, IL-1ß, IL-6, IL-18, PMN elastase and BALF protein levels demonstrate significant lung/systemic inflammation and capillary leak. nCMT-3 significantly ameliorated all of these LPS-induced inflammatory markers to control levels, and decreased the incidence of ALI. CONCLUSIONS: Pre-treatment with nCMT3 significantly attenuates LPS-induced lung injury/inflammation by multiple mechanisms including: MMP activation, PMN elastase, sTREM-1 release, and NLRP3 inflammasome/caspase-1 activation.


Asunto(s)
Lesión Pulmonar Aguda , Proteína con Dominio Pirina 3 de la Familia NLR , Neumonía , Tetraciclinas , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Animales , Antiinflamatorios/farmacología , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Células Precursoras de Granulocitos/metabolismo , Células Precursoras de Granulocitos/patología , Inflamasomas/metabolismo , Inflamación/metabolismo , Interleucina-18/metabolismo , Interleucina-6/metabolismo , Elastasa de Leucocito/metabolismo , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neumonía/tratamiento farmacológico , Neumonía/metabolismo , Neumonía/patología , Tetraciclinas/química , Tetraciclinas/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
5.
J Immunol ; 208(6): 1434-1444, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35246496

RESUMEN

Protein tyrosine phosphatase (PTPase) is critically involved in the regulation of hematopoietic stem cell development and differentiation. Roles of novel isolated receptor PTPase PTPRO from bone marrow hematopoietic stem cells in granulopoiesis have not been investigated. PTPRO expression is correlated with granulocytic differentiation, and Ptpro -/- mice developed neutrophilia, with an expanded granulocytic compartment resulting from a cell-autonomous increase in the number of granulocyte progenitors under steady-state and potentiated innate immune responses against Listeria monocytogenes infection. Mechanistically, mTOR and HIF1α signaling engaged glucose metabolism and initiated a transcriptional program involving the lineage decision factor C/EBPα, which is critically required for the PTPRO deficiency-directed granulopoiesis. Genetic ablation of mTOR or HIF1α or perturbation of glucose metabolism suppresses progenitor expansion, neutrophilia, and higher glycolytic activities by Ptpro -/- In addition, Ptpro -/- upregulated HIF1α regulates the lineage decision factor C/EBPα promoter activities. Thus, our findings identify a previously unrecognized interplay between receptor PTPase PTPRO signaling and mTOR-HIF1α metabolic reprogramming in progenitor cells of granulocytes that underlies granulopoiesis.


Asunto(s)
Células Precursoras de Granulocitos , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores , Animales , Glucosa/metabolismo , Células Precursoras de Granulocitos/metabolismo , Granulocitos/metabolismo , Ratones , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
6.
J Immunol ; 207(4): 1078-1086, 2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-34341172

RESUMEN

Emergency granulopoiesis, also known as demand-adapted granulopoiesis, is defined as the response of an organism to systemic bacterial infections, and it results in neutrophil mobilization from reservoir pools and increased myelopoiesis in the bone marrow. Indirect and direct initiating mechanisms of emergency granulopoiesis have been hypothesized. However, the detailed mechanism of hyperactive myelopoiesis in the bone marrow, which leads to granulocyte left shift, remains unknown. In this study, we report that TLR4 is expressed on granulo-monocytic progenitors, as well as mobilized human peripheral blood CD34+ cells, which account for 0.2% of monocytes in peripheral blood, and ∼ 10% in bone marrow. LPS, a component of Gram-negative bacteria that results in a systemic bacterial infection, induces the differentiation of peripheral blood CD34+ cells into myelocytes and monocytes in vitro via the TLR4 signaling pathway. Moreover, CD34+ cells directly responded to LPS stimulation by activating the MAPK and NF-κB signaling pathways, and they produced IL-6 that promotes emergency granulopoiesis by phosphorylating C/EBPα and C/EBPß, and this effect was suppressed by the action of an IL-6 receptor inhibitor. This work supports the finding that TLR is expressed on human hematopoietic stem and progenitor cells, and it provides evidence that human hematopoietic stem and progenitor cells can directly sense pathogens and produce cytokines exerting autocrine and/or paracrine effects, thereby promoting differentiation.


Asunto(s)
Granulocitos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Interleucina-6/metabolismo , Transducción de Señal/fisiología , Células Madre/metabolismo , Receptor Toll-Like 4/metabolismo , Adaptación Fisiológica/fisiología , Antígenos CD34/metabolismo , Médula Ósea/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular/fisiología , Citocinas/metabolismo , Regulación de la Expresión Génica/fisiología , Células Precursoras de Granulocitos/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Monocitos/metabolismo , Mielopoyesis/fisiología
7.
Development ; 148(17)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34373913

RESUMEN

Neutrophils are the most abundant vertebrate leukocytes and they are essential to host defense. Despite extensive investigation, the molecular network controlling neutrophil differentiation remains incompletely understood. GFI1 is associated with several myeloid disorders, but its role and the role of its co-regulators in granulopoiesis and pathogenesis are far from clear. Here, we demonstrate that zebrafish gfi1aa deficiency induces excessive neutrophil progenitor proliferation, accumulation of immature neutrophils from the embryonic stage, and some phenotypes similar to myelodysplasia syndrome in adulthood. Both genetic and epigenetic analyses demonstrate that immature neutrophil accumulation in gfi1aa-deficient mutants is due to upregulation of cebpa transcription. Increased transcription was associated with Lsd1-altered H3K4 methylation of the cebpa regulatory region. Taken together, our results demonstrate that Gfi1aa, Lsd1 and cebpa form a regulatory network that controls neutrophil development, providing a disease progression-traceable model for myelodysplasia syndrome. Use of this model could provide new insights into the molecular mechanisms underlying GFI1-related myeloid disorders as well as a means by which to develop targeted therapeutic approaches for treatment.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas de Unión al ADN/metabolismo , Hematopoyesis/genética , Histona Demetilasas/metabolismo , Neutrófilos/citología , Proteínas de Pez Cebra/metabolismo , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Diferenciación Celular , Proliferación Celular , Proteínas de Unión al ADN/deficiencia , Embrión no Mamífero , Epigénesis Genética , Células Precursoras de Granulocitos/citología , Células Precursoras de Granulocitos/metabolismo , Histona Demetilasas/genética , Neutrófilos/metabolismo , Pez Cebra , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
8.
Cell Death Dis ; 12(7): 675, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34226527

RESUMEN

Mutations in the transcription factor C/EBPα are found in ~10% of all acute myeloid leukaemia (AML) cases but the contribution of these mutations to leukemogenesis is incompletely understood. We here use a mouse model of granulocyte progenitors expressing conditionally active HoxB8 to assess the cell biological and molecular activity of C/EBPα-mutations associated with human AML. Both N-terminal truncation and C-terminal AML-associated mutations of C/EBPα substantially altered differentiation of progenitors into mature neutrophils in cell culture. Closer analysis of the C/EBPα-K313-duplication showed expansion and prolonged survival of mutant C/EBPα-expressing granulocytes following adoptive transfer into mice. C/EBPα-protein containing the K313-mutation further showed strongly enhanced transcriptional activity compared with the wild-type protein at certain promoters. Analysis of differentially regulated genes in cells overexpressing C/EBPα-K313 indicates a strong correlation with genes regulated by C/EBPα. Analysis of transcription factor enrichment in the differentially regulated genes indicated a strong reliance of SPI1/PU.1, suggesting that despite reduced DNA binding, C/EBPα-K313 is active in regulating target gene expression and acts largely through a network of other transcription factors. Strikingly, the K313 mutation caused strongly elevated expression of C/EBPα-protein, which could also be seen in primary K313 mutated AML blasts, explaining the enhanced C/EBPα activity in K313-expressing cells.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Diferenciación Celular , Células Precursoras de Granulocitos/metabolismo , Leucemia Monocítica Aguda/metabolismo , Mutación , Neutrófilos/metabolismo , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Células Cultivadas , Femenino , Regulación Leucémica de la Expresión Génica , Células Precursoras de Granulocitos/trasplante , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Leucemia Monocítica Aguda/genética , Ratones Endogámicos C57BL , Neutrófilos/trasplante , Regulación hacia Arriba
9.
Methods Mol Biol ; 2182: 117-126, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32894491

RESUMEN

Salmonella enterica is a Gram-negative intracellular pathogen that causes a range of life-threatening diseases in humans and animals worldwide. In a systemic infection, the ability of Salmonella to survive/replicate in macrophages, particularly in the liver and spleen, is crucial for virulence. Transformed macrophage cell lines and primary macrophages prepared from mouse bone marrow are commonly used models for the study of Salmonella infection. However, these models raise technical or ethical issues that highlight the need for alternative methods. This chapter describes a technique for immortalizing early hematopoietic progenitor cells derived from wild-type or transgenic mice and using them to produce macrophages. It validates, through a specific example, the interest of this cellular approach for the study of Salmonella infection.


Asunto(s)
Células Precursoras de Granulocitos/microbiología , Proteínas de Homeodominio/metabolismo , Macrófagos/microbiología , Infecciones por Salmonella/microbiología , Animales , Línea Celular Transformada/metabolismo , Línea Celular Transformada/microbiología , Línea Celular Transformada/patología , Línea Celular Tumoral , Células Precursoras de Granulocitos/metabolismo , Hígado/metabolismo , Hígado/microbiología , Hígado/patología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Salmonella/metabolismo , Infecciones por Salmonella/patología , Salmonella enterica/patogenicidad , Bazo/metabolismo , Bazo/microbiología , Bazo/patología , Virulencia/genética
10.
Life Sci Alliance ; 4(1)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33144337

RESUMEN

Chromosomal rearrangements of the mixed-lineage leukemia gene MLL1 are the hallmark of infant acute leukemia. The granulocyte-macrophage progenitor state forms the epigenetic basis for myelomonocytic leukemia stemness and transformation by MLL-type oncoproteins. Previously, it was shown that the establishment of murine myelomonocytic MLL-ENL transformation, but not its maintenance, depends on the transcription factor C/EBPα, suggesting an epigenetic hit-and-run mechanism of MLL-driven oncogenesis. Here, we demonstrate that compound deletion of Cebpa/Cebpb almost entirely abrogated the growth and survival of MLL-ENL-transformed cells. Rare, slow-growing, and apoptosis-prone MLL-ENL-transformed escapees were recovered from compound Cebpa/Cebpb deletions. The escapees were uniformly characterized by high expression of the resident Cebpe gene, suggesting inferior functional compensation of C/EBPα/C/EBPß deficiency by C/EBPε. Complementation was augmented by ectopic C/EBPß expression and downstream activation of IGF1 that enhanced growth. Cebpe gene inactivation was accomplished only in the presence of complementing C/EBPß, but not in its absence, confirming the Cebpe dependency of the Cebpa/Cebpb double knockouts. Our data show that MLL-transformed myeloid cells are dependent on C/EBPs during the initiation and maintenance of transformation.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/deficiencia , Proteína beta Potenciadora de Unión a CCAAT/deficiencia , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Transformación Celular Neoplásica/genética , Células Precursoras de Granulocitos/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Animales , Apoptosis/genética , Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Ratones , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Fusión Oncogénica/genética , Transducción de Señal/genética , Transfección
11.
RNA ; 27(1): 106-121, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33127860

RESUMEN

Telomeric repeat-containing RNA (TERRA) molecules play important roles at telomeres, from heterochromatin regulation to telomerase activity control. In human cells, TERRA is transcribed from subtelomeric promoters located on most chromosome ends and associates with telomeres. The origin of mouse TERRA molecules is, however, unclear, as transcription from the pseudoautosomal PAR locus was recently suggested to account for the vast majority of TERRA in embryonic stem cells (ESC). Here, we confirm the production of TERRA from both the chromosome 18q telomere and the PAR locus in mouse embryonic fibroblasts, ESC, and various mouse cancer and immortalized cell lines, and we identify two novel sources of TERRA on mouse chromosome 2 and X. Using various approaches, we show that PAR-TERRA molecules account for the majority of TERRA transcripts, displaying an increase of two to four orders of magnitude compared to the telomeric 18q transcript. Finally, we present a SILAC-based pull-down screen revealing a large overlap between TERRA-interacting proteins in human and mouse cells, including PRC2 complex subunits, chromatin remodeling factors, DNA replication proteins, Aurora kinases, shelterin complex subunits, Bloom helicase, Coilin, and paraspeckle proteins. Hence, despite originating from distinct genomic regions, mouse and human TERRA are likely to play similar functions in cells.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , ARN Mensajero/genética , Telómero/química , Transcriptoma , Animales , Aurora Quinasas/genética , Aurora Quinasas/metabolismo , Línea Celular Tumoral , Cromosomas de los Mamíferos/química , Cromosomas de los Mamíferos/metabolismo , Biología Computacional/métodos , Fibroblastos/citología , Fibroblastos/metabolismo , Redes Reguladoras de Genes , Células Precursoras de Granulocitos/citología , Células Precursoras de Granulocitos/metabolismo , Células HeLa , Humanos , Ratones , Monocitos/citología , Monocitos/metabolismo , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Neuronas/citología , Neuronas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , ARN Mensajero/clasificación , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/clasificación , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Complejo Shelterina , Telómero/metabolismo , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo
12.
Int J Mol Sci ; 21(23)2020 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-33255664

RESUMEN

A microfluidic assay for the detection of promyelocytic leukemia (PML)-retinoic acid receptor α (RARα) fusion protein was developed. This microfluidic-based system can be used for rapid personalized differential diagnosis of acute promyelocyte leukemia (APL) with the aim of early initiation of individualized therapy. The fusion protein PML-RARα occurs in 95% of acute promyelocytic leukemia cases and is considered as diagnostically relevant. The fusion protein is formed as a result of translocation t(15,17) and is detected in the laboratory by fluorescence in situ hybridization (FISH) or reverse transcriptase polymerase chain reaction (RT-PCR). Diagnostic methods require many laboratory steps with specialized staff. The developed microfluidic assay includes a sandwich enzyme-linked immunosorbent assay (ELISA) system for PML-RARα on surface of magnetic microparticles in a microfluidic chip. A rapid detection of PML-RARα in cell lysates is achieved in less than one hour. A biotinylated PML-antibody on the surface of magnetic streptavidin coated microparticles is used as capture antibody. The bound translocation product is detected by a RARα antibody conjugated with horseradish peroxidase and the substrate QuantaRed. The analysis is performed in microfluidic channels which involves automated liquid processing with stringent washing and short incubation times. The results of the developed assay show that cell lysates of PML-RARα-positive cells (NB-4) can be clearly distinguished from PML-RARα-negative cells (HL-60, MV4-11).


Asunto(s)
Leucemia Promielocítica Aguda/diagnóstico , Proteínas de Fusión Oncogénica/genética , Proteína de la Leucemia Promielocítica/genética , Receptor alfa de Ácido Retinoico/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/aislamiento & purificación , Células Precursoras de Granulocitos/metabolismo , Células Precursoras de Granulocitos/patología , Humanos , Hibridación Fluorescente in Situ/métodos , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patología , Microfluídica/métodos , Proteínas de Fusión Oncogénica/aislamiento & purificación , Medicina de Precisión , Translocación Genética/genética
13.
JCI Insight ; 5(20)2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32960815

RESUMEN

Giant cell arteritis (GCA) is a common form of primary systemic vasculitis in adults, with no reliable indicators of prognosis or treatment responses. We used single cell technologies to comprehensively map immune cell populations in the blood of patients with GCA and identified the CD66b+CD15+CD10lo/-CD64- band neutrophils and CD66bhiCD15+CD10lo/-CD64+/bright myelocytes/metamyelocytes to be unequivocally associated with both the clinical phenotype and response to treatment. Immature neutrophils were resistant to apoptosis, remained in the vasculature for a prolonged period of time, interacted with platelets, and extravasated into the tissue surrounding the temporal arteries of patients with GCA. We discovered that immature neutrophils generated high levels of extracellular reactive oxygen species, leading to enhanced protein oxidation and permeability of endothelial barrier in an in vitro coculture system. The same populations were also detected in other systemic vasculitides. These findings link functions of immature neutrophils to disease pathogenesis, establishing a clinical cellular signature of GCA and suggesting different therapeutic approaches in systemic vascular inflammation.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Arteritis de Células Gigantes/metabolismo , Neutrófilos/inmunología , Vasculitis Sistémica/inmunología , Enfermedades Vasculares/metabolismo , Anciano , Antígenos CD/metabolismo , Antígenos de Superficie/inmunología , Antígenos de Superficie/metabolismo , Apoptosis/genética , Enfermedades Autoinmunes/sangre , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/patología , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Linaje de la Célula/genética , Técnicas de Cocultivo , Femenino , Proteínas Ligadas a GPI/metabolismo , Arteritis de Células Gigantes/inmunología , Arteritis de Células Gigantes/patología , Células Precursoras de Granulocitos/metabolismo , Células Precursoras de Granulocitos/patología , Humanos , Recuento de Leucocitos , Antígeno Lewis X/metabolismo , Masculino , Persona de Mediana Edad , Neprilisina/metabolismo , Neutrófilos/metabolismo , Neutrófilos/patología , Oxidación-Reducción , Pronóstico , Especies Reactivas de Oxígeno/efectos adversos , Especies Reactivas de Oxígeno/metabolismo , Análisis de la Célula Individual , Vasculitis Sistémica/sangre , Vasculitis Sistémica/metabolismo , Vasculitis Sistémica/patología , Arterias Temporales/inmunología , Arterias Temporales/metabolismo , Arterias Temporales/patología , Enfermedades Vasculares/sangre , Enfermedades Vasculares/inmunología , Enfermedades Vasculares/patología
14.
Exp Hematol ; 85: 20-32.e3, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32437910

RESUMEN

Mds1-Evi1 (also known as Prdm3) and Prdm16 are two highly related zinc finger transcription factors that, within the hematopoietic system, are both expressed primarily in hematopoietic stem cells (HSCs). Our laboratory previously found that constitutive Mds1-Evi1 knockout mice are viable, but their HSCs are unable to withstand myeloablative chemotherapy or effectively transplant irradiated recipient mice. A similar phenotype has been observed for Prdm16, except that the Prdm16 constitutive knockout is lethal. Here, we created a novel double-knockout model of Mds1-Evi1 and Prdm16 in the bone marrow, in which double knockout occurs only in cells that endogenously express Mds1-Evi1 and only upon induction with tamoxifen. We show that combined Mds1-Evi1/Prdm16 deficiency causes bone marrow failure within 15 days, with rapid loss in all progenitor compartments, while the peripheral blood exhibits progressive reductions in peripheral monocytes and granulocytes. We found that surviving hematopoietic stem cells and granulocytic progenitors had elevated apoptosis and cell division, and were unable to form colonies in vitro; adding back wild-type Mds1-Evi1 or Prdm16 to double-knockout bone marrow restores colony formation, and for MDS1-EVI1, this activity depends on a functional PR domain. All of these phenotypic effects were exhibited at milder levels in Mds1-Evi1 and Prdm16 single-knockout controls. Overall, these results illustrate that Mds1-Evi1 and Prdm16 play additive roles in maintaining normal hematopoietic stem cell survival.


Asunto(s)
Apoptosis/fisiología , Proteínas de Unión al ADN/metabolismo , Células Precursoras de Granulocitos/metabolismo , Hematopoyesis/fisiología , Proteína del Locus del Complejo MDS1 y EV11/metabolismo , Modelos Biológicos , Factores de Transcripción/metabolismo , Animales , Línea Celular , Supervivencia Celular/genética , Proteínas de Unión al ADN/genética , Células Precursoras de Granulocitos/citología , Proteína del Locus del Complejo MDS1 y EV11/genética , Ratones , Ratones Noqueados , Factores de Transcripción/genética
15.
Int J Mol Sci ; 21(8)2020 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-32325694

RESUMEN

The intracellular-ubiquitin-ending-enzyme tumor necrosis factor alpha-induced protein 3 (TNFAIP3) is a potent inhibitor of the pro-inflammatory nuclear factor kappa-light-chain-enhancer of activated B cell (NF-kB) pathway. Single nucleotide polymorphisms in TNFAIP3 locus have been associated to autoimmune inflammatory disorders, including Multiple Sclerosis (MS). Previously, we reported a TNFAIP3 down-regulated gene expression level in blood and specifically in monocytes obtained from treatment-naïve MS patients compared to healthy controls (HC). Myeloid cells exert a key role in the pathogenesis of MS. Here we evaluated the effect of specific TNFAIP3 deficiency in myeloid cells including monocytes, monocyte-derived cells (M-MDC) and microglia analyzing lymphoid organs and microglia of mice. TNFAIP3 deletion is induced using conditional knock-out mice for myeloid lineage. Flow-cytometry and histological procedures were applied to assess the immune cell populations of spleen, lymph nodes and bone marrow and microglial cell density in the central nervous system (CNS), respectively. We found that TNFAIP3 deletion in myeloid cells induces a reduction in body weight, a decrease in the number of M-MDC and of common monocyte and granulocyte precursor cells (CMGPs). We also reported that the lack of TNFAIP3 in myeloid cells induces an increase in microglial cell density. The results suggest that TNFAIP3 in myeloid cells critically controls the development of M-MDC in lymphoid organ and of microglia in the CNS.


Asunto(s)
Microglía/citología , Monocitos/citología , Mielopoyesis/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/deficiencia , Animales , Peso Corporal/genética , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Femenino , Citometría de Flujo , Células Precursoras de Granulocitos/citología , Células Precursoras de Granulocitos/metabolismo , Inflamación/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Macrófagos/citología , Macrófagos/inmunología , Masculino , Ratones , Ratones Noqueados , Microglía/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Células Mieloides/citología , Células Mieloides/inmunología , Células Mieloides/metabolismo , Bazo/citología , Bazo/inmunología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo
16.
Nutrition ; 69: 110540, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31525700

RESUMEN

OBJECTIVE: It is well known that protein malnutrition (PM) states can affect hematopoiesis, leading to severe leukopenia and reduced number of granulocytes, which act as the first line of defense, and are important to the innate immune response. The aim of this study was to elucidate some of the mechanisms involved in the impairment of granulopoiesis in PM. METHODS: Male C57BL/6 mice were submitted to PM with a low-protein diet containing 2% protein. Control mice were fed a 12% protein-containing diet. Bone marrow histology and the percentage of granulocytic progenitors were evaluated after in vivo granulocyte-colony stimulating factor (G-CSF) stimulus. Cell proliferation, STAT3 signaling, and the expression of G-CSF receptor were evaluated in hematopoietic progenitor cells. RESULTS: Malnourished animals presented with leukopenia associated with reduced number of granulocytes and reduced percentage of granulocytic progenitors; however, no differences were observed in the regulatory granulopoietic cytokine G-CSF. Additionally, the malnourished group presented with impaired response to in vivo G-CSF stimulus compared with control animals. PM was implicated in decreased ability of c-Kit+ cells to differentiate into myeloid progenitor cells and downregulated STAT3 signaling. Furthermore, the malnourished group exhibited reduced expression of G-CSF receptor on granule-monocytic progenitors. This reduced expression was not completely reversible with G-CSF treatment. CONCLUSIONS: This study implies that PM promotes intrinsic alterations to hematopoietic precursors, which result in hematologic changes, mainly neutropenia, observed in peripheral blood in PM states.


Asunto(s)
Dieta con Restricción de Proteínas/efectos adversos , Células Precursoras de Granulocitos/metabolismo , Neutropenia/sangre , Deficiencia de Proteína/sangre , Receptores de Factor Estimulante de Colonias de Granulocito/sangre , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Neutropenia/etiología , Deficiencia de Proteína/etiología
17.
Int J Mol Sci ; 20(22)2019 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-31717318

RESUMEN

Granulocyte-colony stimulating factor (G-CSF)/nicotinamide phosphoribosyltransferase (NAMPT) signaling has been shown to be crucial for the modulation of neutrophil development and functionality. As this signaling pathway is significantly suppressed by type I interferons (IFNs), we aimed to study how the regulation of neutrophil differentiation and phenotype is altered in IFN-deficient mice during granulopoiesis. The composition of bone marrow granulocyte progenitors and their Nampt expression were assessed in bone marrow of type I IFN receptor knockout (Ifnar1-/-) mice and compared to wild-type animals. The impact of NAMPT inhibition on the proliferation, survival, and differentiation of murine bone marrow progenitors, as well as of murine 32D and human HL-60 neutrophil-like cell lines, was estimated. The progressive increase of Nampt expression during neutrophil progenitor maturation could be observed, and it was more prominent in IFN-deficient animals. Altered composition of bone marrow progenitors in these mice correlated with the dysregulation of apoptosis and altered differentiation of these cells. We observed that NAMPT is vitally important for survival of early progenitors, while at later stages it delays the differentiation of neutrophils, with moderate effect on their survival. This study shows that IFN-deficiency leads to the elevated NAMPT expression in the bone marrow, which in turn modulates neutrophil development and differentiation, even in the absence of tumor-derived stimuli.


Asunto(s)
Diferenciación Celular , Interferones/metabolismo , Neutrófilos/citología , Neutrófilos/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Transducción de Señal , Animales , Apoptosis , Supervivencia Celular , Factor Estimulante de Colonias de Granulocitos/metabolismo , Células Precursoras de Granulocitos/metabolismo , Células HL-60 , Humanos , Ratones Endogámicos C57BL , Infiltración Neutrófila , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/metabolismo
18.
PLoS One ; 14(9): e0223013, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31568521

RESUMEN

INTRODUCTION: Vacuolization is a frequently found morphological feature in acute myeloid leukemia (AML) blasts. Subcellular origin and biological function as well as prognostic impact are currently unknown. The aim of this study was to evaluate whether vacuolization correlates with clinically relevant AML features. MATERIALS & METHODS: Bone marrow smears of patients diagnosed with AML at the University Hospital Frankfurt between January 2011 and August 2013 were analyzed for blast vacuolization and correlated with clinically relevant AML features. Patients undergoing standard induction chemotherapy were further analyzed for molecular and cytogenetic features as well as treatment response and survival. RESULTS: 14 of 100 patients diagnosed with AML receiving standard induction chemotherapy had evidence of blast vacuolization. Positivity for vacuolization correlated with a CD15 positive immunophenotype and with a higher incidence of high-risk AML according to the European LeukemiaNet risk stratification. AML patients with blast vacuolization had a poor blast clearance after standard induction chemotherapy and poor survival. DISCUSSION: In conclusion, our findings demonstrate that vacuolization can easily be determined in myeloid leukemia blasts and may be a useful biomarker to predict AML risk groups as well as early treatment response rates and survival.


Asunto(s)
Médula Ósea/patología , Células Precursoras de Granulocitos/patología , Quimioterapia de Inducción/métodos , Leucemia Mieloide Aguda/diagnóstico , Vacuolas/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Médula Ósea/metabolismo , Médula Ósea/ultraestructura , Femenino , Células Precursoras de Granulocitos/metabolismo , Células Precursoras de Granulocitos/ultraestructura , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Antígeno Lewis X/genética , Antígeno Lewis X/inmunología , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Medición de Riesgo , Análisis de Supervivencia , Vacuolas/metabolismo , Vacuolas/ultraestructura
19.
J Immunotoxicol ; 16(1): 63-73, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31282784

RESUMEN

Engineered nanomaterials (ENM) are being used in a wide range of consumer products and pharmaceuticals; hence, there is an increasing risk for human exposure and potential adverse outcomes. The immune system, vital in host defense and protection against environmental agents, is typically initiated and executed by innate effector immune cells including macrophages and neutrophils. Previous literature has reported the immune system as a major target of ENM toxicity; however, there is inconsistency regarding the immunotoxicity of ENM. This could be attributed to differences in ENM physicochemical properties, cellular models examined, biocorona formation, etc. Thus, the current study examined the toxicity and immunomodulatory effects of silver nanoparticles (AgNP), one of the most utilized ENM in consumer and medical products, in two key innate immune cell models, e.g. RAW 264.7 cells (macrophages) and differentiated MPRO 2.1 cells (promyelocytes/neutrophils). The results showed that despite a generation of reactive oxygen species, exposure to 20 nm citrate-coated AgNP was not associated with major oxidative damage, inflammatory responses, nor cytotoxicity. Nevertheless, and most importantly, pre-exposure to the AgNP for 24 h enhanced RAW 264.7 cell phagocytic ability as well as the release of inflammatory cytokine interleukin-6 in response to lipopolysaccharide (LPS). In MPRO 2.1 cells, AgNP pre-exposure also resulted in enhanced phagocytic ability; however, these cells manifest reduced cell degranulation (elastase release) and oxidative burst in response to phorbol myristate acetate (PMA). Taken together, these findings indicated to us that exposure to AgNP, despite not being directly (cyto)toxic to these cells, had the potential to alter immune cell responses. The findings underscore the import of assessing immune cell function post-exposure to ENM beyond the standard endpoints such as oxidative stress and cytotoxicity. In addition, these findings further illustrate the importance of understanding the underlying molecular mechanisms of ENM-cellular interactions, particularly in the immune system.


Asunto(s)
Células Precursoras de Granulocitos/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Neutrófilos/efectos de los fármacos , Plata/toxicidad , Animales , Degranulación de la Célula/efectos de los fármacos , Degranulación de la Célula/inmunología , Células Precursoras de Granulocitos/inmunología , Células Precursoras de Granulocitos/metabolismo , Interleucina-6/inmunología , Interleucina-6/metabolismo , Lipopolisacáridos/inmunología , Ratones , Neutrófilos/inmunología , Neutrófilos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/inmunología , Tamaño de la Partícula , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Células RAW 264.7 , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Pruebas de Toxicidad
20.
Exp Cell Res ; 382(1): 111445, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31152707

RESUMEN

MicroRNAs (miRNA) are small noncoding RNAs that regulate gene expression by targeting mRNAs in a sequence specific manner, thereby determining their degradation or inhibiting translation. They are involved in processes such as proliferation, differentiation and apoptosis by fine-tuning the expression of genes underlying such events. The expression of specific miRNAs is involved in hematopoietic differentiation and their deregulation contributes to the development of hematopoietic malignancies such as acute myeloid leukemia (AML). miR-130a is over-expressed in AML. Here we show that miR-130a is physiologically expressed in myeloblasts and down-regulated during monocyte differentiation. Gain- and loss-of-function experiments performed on CD34+ human hematopoietic stem cells confirmed that expression of miR-130a inhibits monocyte differentiation by interfering with the expression of key transcription factors HOXA10, IRF8, KLF4, MAFB and PU-1. The data obtained in this study highlight that the correct modulation of miR-130a is necessary for normal differentiation to occur and confirming that deregulation of this miRNA might underlie the differentiation block occurring in AML.


Asunto(s)
Regulación de la Expresión Génica , Células Precursoras de Granulocitos/metabolismo , Células Madre Hematopoyéticas/metabolismo , MicroARNs/fisiología , Monocitos/citología , Mielopoyesis/fisiología , Proteínas de Neoplasias/fisiología , Antígenos CD34/análisis , Línea Celular Tumoral , Linaje de la Célula , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Mutación con Ganancia de Función , Células Precursoras de Granulocitos/citología , Células Madre Hematopoyéticas/citología , Humanos , Factor 4 Similar a Kruppel , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Mutación con Pérdida de Función , MicroARNs/antagonistas & inhibidores , MicroARNs/biosíntesis , MicroARNs/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Ácidos Nucleicos de Péptidos/farmacología , ARN Neoplásico/genética , ARN Neoplásico/fisiología , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA