Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 514
Filter
1.
Nat Immunol ; 18(8): 877-888, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28650480

ABSTRACT

The origin and specification of human dendritic cells (DCs) have not been investigated at the clonal level. Through the use of clonal assays, combined with statistical computation, to quantify the yield of granulocytes, monocytes, lymphocytes and three subsets of DCs from single human CD34+ progenitor cells, we found that specification to the DC lineage occurred in parallel with specification of hematopoietic stem cells (HSCs) to the myeloid and lymphoid lineages. This started as a lineage bias defined by specific transcriptional programs that correlated with the combinatorial 'dose' of the transcription factors IRF8 and PU.1, which was transmitted to most progeny cells and was reinforced by upregulation of IRF8 expression driven by the hematopoietic cytokine FLT3L during cell division. We propose a model in which specification to the DC lineage is driven by parallel and inheritable transcriptional programs in HSCs and is reinforced over cell division by recursive interactions between transcriptional programs and extrinsic signals.


Subject(s)
Cell Lineage , Dendritic Cells/cytology , Hematopoietic Stem Cells/cytology , Interferon Regulatory Factors/metabolism , Leukopoiesis , Multipotent Stem Cells/cytology , Animals , Cell Differentiation , Fetal Blood , Flow Cytometry , Humans , Interferon Regulatory Factors/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Principal Component Analysis , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Up-Regulation
2.
PLoS Genet ; 17(8): e1009693, 2021 08.
Article in English | MEDLINE | ID: mdl-34351909

ABSTRACT

The ubiquitin-proteasome system plays important roles in various biological processes as it degrades the majority of cellular proteins. Adequate proteasomal degradation of crucial transcription regulators ensures the proper development of neutrophils. The ubiquitin E3 ligase of Growth factor independent 1 (GFI1), a key transcription repressor governing terminal granulopoiesis, remains obscure. Here we report that the deficiency of the ring finger protein Interferon regulatory factor 2 binding protein 2a (Irf2bp2a) leads to an impairment of neutrophils differentiation in zebrafish. Mechanistically, Irf2bp2a functions as a ubiquitin E3 ligase targeting Gfi1aa for proteasomal degradation. Moreover, irf2bp2a gene is repressed by Gfi1aa, thus forming a negative feedback loop between Irf2bp2a and Gfi1aa during neutrophils maturation. Different levels of GFI1 may turn it into a tumor suppressor or an oncogene in malignant myelopoiesis. Therefore, discovery of certain drug targets GFI1 for proteasomal degradation by IRF2BP2 might be an effective anti-cancer strategy.


Subject(s)
DNA-Binding Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Zebrafish Proteins/metabolism , Zebrafish/growth & development , Animals , DNA-Binding Proteins/metabolism , Feedback, Physiological , Female , Gene Expression Regulation , Gene Knockout Techniques , HEK293 Cells , HL-60 Cells , Humans , Leukopoiesis , Male , Proteolysis , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
4.
Blood ; 136(12): 1394-1401, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32730592

ABSTRACT

Vitamin C serves as a cofactor for Fe(II) and 2-oxoglutarate-dependent dioxygenases including TET family enzymes, which catalyze the oxidation of 5-methylcytosine into 5-hydroxymethylcytosine and further oxidize methylcytosines. Loss-of-function mutations in epigenetic regulators such as TET genes are prevalent in hematopoietic malignancies. Vitamin C deficiency is frequently observed in cancer patients. In this review, we discuss the role of vitamin C and TET proteins in cancer, with a focus on hematopoietic malignancies, T regulatory cells, and other immune system cells.


Subject(s)
Ascorbic Acid/immunology , Dioxygenases/immunology , Immunity , Neoplasms/immunology , Animals , Ascorbic Acid Deficiency/complications , Ascorbic Acid Deficiency/immunology , Ascorbic Acid Deficiency/physiopathology , Humans , Ketoglutaric Acids/immunology , Leukopoiesis , Neoplasms/complications , Neoplasms/physiopathology , T-Lymphocytes, Regulatory/immunology
5.
J Immunol ; 205(10): 2786-2794, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32998984

ABSTRACT

BP180 (also termed type XVII collagen) is a hemidesmosomal protein and plays a critical role in cell-cell matrix adhesion in the skin; however, its other biological functions are largely unclear. In this study, we generated a BP180 functional-deficient mouse strain by deleting its extracellular domain of humanized NC16A (termed ΔNC16A mice). We found that BP180 is expressed by bone marrow mesenchymal stem cells (BM-MSC), and its functional deficiency leads to myeloid hyperplasia. Altered granulopoiesis in ΔNC16A mice is through bone marrow stromal cells evidenced by bone marrow transplantation. Furthermore, the level of G-CSF in bone marrow and circulation were significantly increased in ΔNC16A mice as compared with wild-type mice. The increased G-CSF was accompanied by an increased activation of the NF-κB signaling pathway in bone marrow and BM-MSC of ΔNC16A mice. Blockade of G-CSF restored normal granulopoiesis in ΔNC16A mice. Inhibition of NF-κB signaling pathway significantly reduces the release of G-CSF from ΔNC16A BM-MSC in vitro and the level of serum G-CSF in ΔNC16A mice. To our knowledge, these findings provide the first direct evidence that BP180 plays an important role in granulopoiesis through regulating NF-κB signaling pathway in BM-MSC.


Subject(s)
Autoantigens/metabolism , Bone Marrow/pathology , Leukopoiesis/immunology , Mesenchymal Stem Cells/metabolism , Neutrophils/physiology , Non-Fibrillar Collagens/metabolism , Animals , Autoantigens/genetics , Bone Marrow/drug effects , Bone Marrow/metabolism , Cell Differentiation/immunology , Disease Models, Animal , Granulocyte Colony-Stimulating Factor/antagonists & inhibitors , Granulocyte Colony-Stimulating Factor/blood , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Hyperplasia/genetics , Hyperplasia/immunology , Mice , Mice, Transgenic , NF-kappa B/metabolism , Non-Fibrillar Collagens/genetics , Protein Domains/genetics , Signal Transduction/drug effects , Signal Transduction/immunology , Collagen Type XVII
6.
J Biol Chem ; 295(28): 9663-9675, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32467231

ABSTRACT

Acute myeloid leukemia (AML) with mixed lineage leukemia 1 (MLL1) gene rearrangement is characterized by increased expression of a set of homeodomain transcription factors, including homeobox A9 (HOXA9) and HOXA10. The target genes for these regulators include fibroblast growth factor 2 (FGF2) and Ariadne RBR E3 ubiquitin ligase 2 (ARIH2). FGF2 induces leukemia stem cell expansion in MLL1-rearranged AML. ARIH2 encodes TRIAD1, an E3 ubiquitin ligase required for termination of emergency granulopoiesis and leukemia suppressor function in MLL1-rearranged AML. Receptor tyrosine kinases (RTKs), including the FGF receptor, are TRIAD1 substrates that are possibly relevant to these activities. Using transcriptome analysis, we found increased activity of innate immune response pathways and RTK signaling in bone marrow progenitors from mice with MLL1-rearranged AML. We hypothesized that sustained RTK signaling, because of decreased TRIAD1 activity, impairs termination of emergency granulopoiesis during the innate immune response and contributes to leukemogenesis in this AML subtype. Consistent with this, we found aberrantly sustained emergency granulopoiesis in a murine model of MLL1-rearranged AML, associated with accelerated leukemogenesis. Treating these mice with an inhibitor of TRIAD1-substrate RTKs terminated emergency granulopoiesis, delayed leukemogenesis during emergency granulopoiesis, and normalized innate immune responses when combined with chemotherapy. Emergency granulopoiesis also hastened postchemotherapy relapse in mice with MLL1-rearranged AML, but remission was sustained by ongoing RTK inhibition. Our findings suggest that the physiological stress of infectious challenges may drive AML progression in molecularly defined subsets and identify RTK inhibition as a potential therapeutic approach to counteract this process.


Subject(s)
Gene Rearrangement , Histone-Lysine N-Methyltransferase/metabolism , Leukemia, Myeloid, Acute/enzymology , Leukopoiesis , Myeloid-Lymphoid Leukemia Protein/metabolism , Neoplasms, Experimental/enzymology , Animals , ErbB Receptors/genetics , ErbB Receptors/metabolism , Histone-Lysine N-Methyltransferase/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplastic Stem Cells/pathology , Recurrence , Signal Transduction/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
7.
Blood ; 134(14): 1159-1175, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31366618

ABSTRACT

Hematopoietic transcription factor LIM domain only 2 (LMO2), a member of the TAL1 transcriptional complex, plays an essential role during early hematopoiesis and is frequently activated in T-cell acute lymphoblastic leukemia (T-ALL) patients. Here, we demonstrate that LMO2 is activated by deacetylation on lysine 74 and 78 via the nicotinamide phosphoribosyltransferase (NAMPT)/sirtuin 2 (SIRT2) pathway. LMO2 deacetylation enables LMO2 to interact with LIM domain binding 1 and activate the TAL1 complex. NAMPT/SIRT2-mediated activation of LMO2 by deacetylation appears to be important for hematopoietic differentiation of induced pluripotent stem cells and blood formation in zebrafish embryos. In T-ALL, deacetylated LMO2 induces expression of TAL1 complex target genes HHEX and NKX3.1 as well as LMO2 autoregulation. Consistent with this, inhibition of NAMPT or SIRT2 suppressed the in vitro growth and in vivo engraftment of T-ALL cells via diminished LMO2 deacetylation. This new molecular mechanism may provide new therapeutic possibilities in T-ALL and may contribute to the development of new methods for in vitro generation of blood cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Hematopoiesis , LIM Domain Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Acetylation , Animals , Cells, Cultured , HEK293 Cells , Humans , Leukopoiesis , Mice , Models, Molecular , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Zebrafish
8.
Bull Exp Biol Med ; 172(2): 195-201, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34855087

ABSTRACT

We analyzed advantages of the liposomal form of Xymedon (50 and 100 mg/kg) over free Xymedon (in the corresponding doses) in leukopoiesis restoration in rats with Walker-256 carcinoma treated with liposomal combination of doxorubicin (4 mg/kg) and cyclophosphamide (45 mg/kg) (single intravenous injection on day 11 after transplantation of tumor cells). Liposomal and free Xymedon were injected intravenously over 5 days starting from day 11 of the experiment. Changes in leukopoiesis in peripheral blood and myelograms were assessed on days 3 and 7 after chemotherapy. Liposomal Xymedon in both doses (unlike its free form) 2-fold increased the number of lymphocytes on day 3 after chemotherapy in comparison with the level observed after administration of liposomal cytostatics alone. Liposomal Xymedon in a dose of 50 mg/kg (but not 100 mg/kg) promoted the maintenance of monocyte count at the level of intact control on days 3 and 7 after chemotherapy. Liposomal Xymedon in a dose of 50 mg/kg and free Xymedon in a dose of 100 mg/kg equally stimulated the increase in myelocytes content in the bone marrow to the level of intact control on day 3 after chemotherapy, thus promoting restoration of granulocytopoiesis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Leukopoiesis/drug effects , Pyrimidines/administration & dosage , Animals , Carcinoma 256, Walker/drug therapy , Carcinoma 256, Walker/pathology , Cyclophosphamide/administration & dosage , Dosage Forms , Doxorubicin/administration & dosage , Female , Leukopoiesis/physiology , Liposomes/administration & dosage , Myeloablative Agonists/therapeutic use , Rats , Rats, Wistar
9.
Semin Cell Dev Biol ; 75: 23-39, 2018 03.
Article in English | MEDLINE | ID: mdl-28866328

ABSTRACT

Splicing is a ubiquitous process in eukaryotic cells, long recognised as contributing to diversity of the transcriptome. More specifically, splicing fine-tunes the transcriptome output for highly individual outcomes at different stages of cell development, in specific timeframes, which when perturbed result in significant human diseases. Granulopoiesis provides a particularly well studied example of how splicing can be a highly flexible but tightly regulated process. Focusing on the specific case of granulopoiesis, this review surveys the contribution of cis-splicing variations in individual genes and the trans-regulation of global splicing outcomes during the normal development of neutrophils. Further, the contribution of splicing dysfunction to the pathogenesis of diseases of neutrophil number, function and maturation including hereditary neutropenia, myelodysplasia, and acute myeloid leukaemia is explored.


Subject(s)
Alternative Splicing , Leukopoiesis/genetics , Neutropenia/genetics , Neutrophils/metabolism , Animals , Gene Expression Regulation , Genetic Predisposition to Disease/genetics , Humans , Models, Genetic , Mutation
10.
Blood ; 142(6): 505-507, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37561541
11.
Blood ; 132(12): 1265-1278, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30064973

ABSTRACT

Genetic studies have identified recurrent somatic mutations in acute myeloid leukemia (AML) patients, including in the Wilms' tumor 1 (WT1) gene. The molecular mechanisms by which WT1 mutations contribute to leukemogenesis have not yet been fully elucidated. We investigated the role of Wt1 gene dosage in steady-state and pathologic hematopoiesis. Wt1 heterozygous loss enhanced stem cell self-renewal in an age-dependent manner, which increased stem cell function over time and resulted in age-dependent leukemic transformation. Wt1-haploinsufficient leukemias were characterized by progressive genetic and epigenetic alterations, including those in known leukemia-associated alleles, demonstrating a requirement for additional events to promote hematopoietic transformation. Consistent with this observation, we found that Wt1 depletion cooperates with Flt3-ITD mutation to induce fully penetrant AML. Our studies provide insight into mechanisms of Wt1-loss leukemogenesis and into the evolutionary events required to induce transformation of Wt1-haploinsufficient stem/progenitor cells.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Mutation , Repressor Proteins/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Self Renewal , Gene Deletion , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Leukemia, Myeloid, Acute/pathology , Leukopoiesis , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Myeloid Cells/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , WT1 Proteins , fms-Like Tyrosine Kinase 3/genetics
12.
Pharm Res ; 37(6): 102, 2020 May 21.
Article in English | MEDLINE | ID: mdl-32440783

ABSTRACT

PURPOSE: To model absolute neutrophil count (ANC) suppression in response to acute radiation (AR) exposure and evaluate ANC time course as a predictor of overall survival (OS) in response to AR exposure with or without treatment with granulocyte colony-stimulating factor in nonhuman primates. METHODS: Source data were obtained from two pivotal studies conducted in rhesus macaques exposed to 750 cGy of whole body irradiation on day 0 that received either placebo, daily filgrastim, or pegfilgrastim (days 1 and 8 after irradiation). Animals were observed for 60 days with ANC measured every 1 to 2 days. The population model of ANC response to AR and the link between observed ANC time course and OS consisted of three submodels characterizing injury due to radiation, granulopoiesis, and a time-to-event model of OS. RESULTS: The ANC response model accurately described the effects of AR exposure on the duration of neutropenia. ANC was a valid surrogate for survival because it explained 76% (95% CI, 41%-97%) and 73.2% (95% CI, 38.7%-99.9%) of the treatment effect for filgrastim and pegfilgrastim, respectively. CONCLUSION: The current model linking radiation injury to neutropenia and ANC time course to OS can be used as a basis for translating these effects to humans.


Subject(s)
Filgrastim/administration & dosage , Models, Biological , Neutropenia/prevention & control , Neutrophils , Polyethylene Glycols/administration & dosage , Radiation Injuries, Experimental/prevention & control , Animals , Feasibility Studies , Female , Leukocyte Count , Leukopoiesis/drug effects , Leukopoiesis/radiation effects , Macaca mulatta , Male , Neutropenia/blood , Neutropenia/etiology , Neutropenia/mortality , Radiation Injuries, Experimental/blood , Radiation Injuries, Experimental/mortality , Time Factors
13.
J Immunol ; 200(6): 2129-2139, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29427417

ABSTRACT

Emergency (stress) granulopoiesis is an episodic process for the production of granulocytes in response to infectious challenge. We previously determined that Fanconi C, a component of the Fanconi DNA-repair pathway, is necessary for successful emergency granulopoiesis. Fanconi anemia results from mutation of any gene in this pathway and is characterized by bone marrow failure (BMF) in childhood and clonal progression in adolescence. Although murine Fanconi anemia models exhibit relatively normal steady-state hematopoiesis, FANCC-/- mice are unable to mount an emergency granulopoiesis response. Instead, these mice develop BMF and die during repeated unsuccessful emergency granulopoiesis attempts. In FANCC-/- mice, BMF is associated with extensive apoptosis of hematopoietic stem and progenitor cells through an undefined mechanism. In this study, we find that TP53 haploinsufficiency completely rescues emergency granulopoiesis in FANCC-/- mice and protects them from BMF during repeated emergency granulopoiesis episodes. Instead, such recurrent challenges accelerated clonal progression in FANCC-/-TP53+/- mice. In FANCC-/- mice, BMF during multiple emergency granulopoiesis attempts was associated with increased ataxia telangiectasia and Rad3-related protein (Atr) and p53 activation with each attempt. In contrast, we found progressive attenuation of expression and activity of Atr, and consequent p53 activation and apoptosis, in the bone marrow of FANCC-/-TP53+/- mice during this process. Therefore, activation of Atr-with consequent Fanconi-mediated DNA repair or p53-dependent apoptosis-is an essential component of emergency granulopoiesis and it protects the bone marrow from genotoxic stress during this process.


Subject(s)
Fanconi Anemia Complementation Group C Protein/metabolism , Granulocytes/metabolism , Haploinsufficiency/physiology , Leukopoiesis/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/physiology , Ataxia Telangiectasia Mutated Proteins/metabolism , Bone Marrow/metabolism , DNA Damage/physiology , DNA Repair/physiology , Fanconi Anemia/metabolism , Mice
14.
PLoS Genet ; 13(10): e1007027, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28976973

ABSTRACT

MicroRNAs are universal post-transcriptional regulators in genomes. They have the ability of buffering gene expressional programs, contributing to robustness of biological systems and playing important roles in development, physiology and diseases. Here, we identified a microRNA, miR-125a, as a positive regulator of granulopoiesis. MiR125a knockout mice show reduced infiltration of neutrophils in the lung and alleviated tissue destruction after endotoxin challenge as a consequence of decreased neutrophil numbers. Furthermore, we demonstrated that this significant reduction of neutrophils was due to impaired development of granulocyte precursors to mature neutrophils in an intrinsic manner. We showed that Socs3, a critical repressor for granulopoiesis, was a target of miR-125a. Overall, our study revealed a new microRNA regulating granulocyte development and supported a model in which miR-125a acted as a fine-tuner of granulopoiesis.


Subject(s)
Leukopoiesis/genetics , Leukopoiesis/physiology , MicroRNAs/genetics , MicroRNAs/metabolism , Neutrophils/cytology , Neutrophils/metabolism , 3' Untranslated Regions , Animals , Binding Sites/genetics , Cell Death , Cell Differentiation , Cell Proliferation , Granulocyte Colony-Stimulating Factor/metabolism , Granulocytes/cytology , Granulocytes/metabolism , Mice , Mice, Knockout , Models, Biological , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Shock, Septic/genetics , Shock, Septic/metabolism , Shock, Septic/pathology , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism
15.
Immunity ; 33(4): 620-31, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-21029969

ABSTRACT

Although carcinoembryonic antigen-related cell adhesion molecule-1 (CEACAM1) is an activation marker for neutrophils and delays neutrophil apoptosis, the role of CEACAM1 in granulopoiesis and neutrophil-dependent host immune responses has not been investigated. CEACAM1 expression correlated with granulocytic differentiation, and Ceacam1(-/-) mice developed neutrophilia because of loss of the Src-homology-phosphatase-1 (SHP-1)-dependent inhibition of granulocyte colony-stimulating factor receptor (G-CSFR) signal transducer and activator of transcription (Stat3) pathway provided by CEACAM1. Moreover, Ceacam1(-/-) mice were hypersensitive to Listeria Monocytogenes (LM) infection with an accelerated mortality. Reintroduction of CEACAM1 into Ceacam1(-/-) bone marrow restored normal granulopoiesis and host sensitivity to LM infection, while mutation of its immunoreceptor tyrosine-based inhibitory motifs (ITIMs) abrogated this restoration. shRNA-mediated reduction of Stat3 amounts rescued normal granulopoiesis, attenuating host sensitivity to LM infection in Ceacam1(-/-) mice. Thus, CEACAM1 acted as a coinhibitory receptor for G-CSFR regulating granulopoiesis and host innate immune response to bacterial infections.


Subject(s)
Carcinoembryonic Antigen/physiology , Granulocytes/physiology , Leukopoiesis , Receptors, Granulocyte Colony-Stimulating Factor/antagonists & inhibitors , Animals , Cell Lineage , Cell Proliferation , Mice , Mice, Inbred C57BL , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , RNA, Small Interfering/genetics , STAT3 Transcription Factor/physiology , Signal Transduction
16.
Phytother Res ; 33(2): 452-460, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30548344

ABSTRACT

The milk thistle compound Silibinin (i.e., a 1:1 mixture of Silybin A and Silybin B) stimulates vasculogenesis of mouse embryonic stem (ES) cells. Because vasculogenesis and leukopoiesis are interrelated, the effect of Silibinin on leukopoiesis of ES cells was investigated. Treatment of differentiating ES cells with hydrosoluble Silibinin-C-2',3-dihydrogen succinate dose-dependent increased the number of CD18+ , CD45+ , and CD68+ cells, indicating leukocyte/macrophage differentiation. Silibinin treatment activated phosphoinositide 3-kinase (PI3K), AKT (protein kinase B), signal transducer and activator of transcription 3 (STAT3), stimulated hypoxia-induced factor-1α (HIF-1α), and vascular endothelial growth factor receptor 2 (VEGFR2) expression and raised intracellular nitric oxide (NO). Western blot experiments showed that upon coincubation with either the PI3K inhibitor LY294002, the STAT3 inhibitor Stattic, the AKT antagonist AKT inhibitor VIII, or the NO inhibitor L-NAME, the Silibinin-induced expression of CD18, CD45, and CD68 was abolished. Moreover, the stimulation of HIF-1α and VEGFR2 expression was blunted upon STAT3 and PI3K/AKT inhibition. Treatment of differentiating ES cells with L-NAME abolished the stimulation of VEGFR2 and VE-cadherin expression achieved with Silibinin, indicating that NO is involved in vasculogenesis and leukocyte differentiation pathways. In summary, the data of the present study demonstrate that Silibinin stimulates leukocyte differentiation of ES cells, which is associated to vasculogenesis and regulated by PI3K/AKT-, STAT3-, and NO-mediated signaling.


Subject(s)
Leukopoiesis/drug effects , Mouse Embryonic Stem Cells/drug effects , Silybin/pharmacology , Silybum marianum/chemistry , Animals , Chromones/pharmacology , Mice , Morpholines/pharmacology , Nitric Oxide/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism
17.
Bull Exp Biol Med ; 168(2): 275-279, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31782001

ABSTRACT

We studied the effects of combined chemotherapy with doxorubicin/docetaxel on erythroid and granulocytic hematopoietic lineages with particular attention focused on their recovery in patients with stages III-IV breast cancer. Intensification of differentiation of erythroid and granulocytic CFU (even under conditions of their suppressed proliferation) provided the increase in the content of mature and morphologically differentiated elements in the bone marrow and peripheral blood. High proliferative activity of erythroid and granulomonocytic precursors resulted from enhanced production of hematopoiesis-stimulating activities by microenvironment elements.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Docetaxel/therapeutic use , Doxorubicin/therapeutic use , Erythropoiesis/drug effects , Leukopoiesis/drug effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow/metabolism , Cell Lineage/drug effects , Erythrocytes/cytology , Female , Granulocyte Colony-Stimulating Factor/metabolism , Granulocytes/cytology , Humans
18.
Bull Exp Biol Med ; 166(3): 344-347, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30627910

ABSTRACT

The role of signaling molecules in synthesis of humoral regulators of granulocytopoiesis by the hematopoietic microenvironmental cells during stress was analyzed using specific inhibitors. The major role in stimulation of the synthesis of granulocytic CSF during stressful stimulation is played by PI3K/Akt signaling cascade. Nuclear transcription factor NF-κB plays an auxiliary role in the regulation of functional activity of the bone marrow mononuclears. However, this factor affects the synthesis of granulocytic CSF by CD4+ cells of the bone marrow in response to stressful stimulation. Different degree and specific character of involvement of the signaling proteins in the regulation of the production of humoral factors determining colony-stimulating activity are explained by changes in functional state of monocyte-derived macrophages in different periods of stress response.


Subject(s)
Granulocyte Colony-Stimulating Factor/genetics , Granulocytes/immunology , NF-kappa B/genetics , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction/immunology , Stress, Psychological/genetics , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Chromones/pharmacology , Flavonoids/pharmacology , Gene Expression Regulation , Gold Sodium Thiomalate/pharmacology , Granulocyte Colony-Stimulating Factor/immunology , Granulocytes/drug effects , Granulocytes/pathology , Imidazoles/pharmacology , Immobilization/methods , Leukopoiesis/drug effects , Leukopoiesis/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Morpholines/pharmacology , NF-kappa B/antagonists & inhibitors , NF-kappa B/immunology , Phosphatidylinositol 3-Kinases/immunology , Phosphoinositide-3 Kinase Inhibitors , Pyridines/pharmacology , Stress, Psychological/immunology , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
19.
Biochem Biophys Res Commun ; 498(3): 466-472, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29501745

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) receptor (G-CSFR) is a type I cytokine receptor which is involved in hematopoietic cell maturation. G-CSFR has three putative C-mannosylation sites at W253, W318, and W446; however, it is not elucidated whether G-CSFR is C-mannosylated or not. In this study, we first demonstrated that G-CSFR was C-mannosylated at only W318. We also revealed that C-mannosylation of G-CSFR affects G-CSF-dependent downstream signaling through changing ligand binding capability but not cell surface localization. Moreover, C-mannosylation of G-CSFR was functional and regulated granulocytic differentiation in myeloid 32D cells. In conclusion, we found that G-CSFR is C-mannosylated at W318 and that this C-mannosylation has role(s) for myeloid cell differentiation through regulating downstream signaling.


Subject(s)
Granulocytes/cytology , Leukopoiesis , Mannose/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Amino Acid Sequence , Cell Line, Tumor , Granulocytes/metabolism , HEK293 Cells , Humans , Mannose/analysis , Receptors, Granulocyte Colony-Stimulating Factor/chemistry , Signal Transduction
20.
Am J Physiol Regul Integr Comp Physiol ; 315(2): R218-R229, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29641231

ABSTRACT

The interaction between C-X-C chemokine receptor type 4 (CXCR4) and its cognate ligand C-X-C motif chemokine ligand 12 (CXCL12) plays a critical role in regulating hematopoietic stem cell activation and subsequent cellular mobilization. Extensive studies of these genes have been conducted in mammals, but much less is known about the expression and function of CXCR4 and CXCL12 in non-mammalian vertebrates. In the present study, we identify simultaneous expression of CXCR4 and CXCL12 orthologs in the epigonal organ (the primary hematopoietic tissue) of the little skate, Leucoraja erinacea. Genetic and phylogenetic analyses were functionally supported by significant mobilization of leukocytes following administration of Plerixafor, a CXCR4 antagonist and clinically important drug. Our results provide evidence that, as in humans, Plerixafor disrupts CXCR4/CXCL12 binding in the little skate, facilitating release of leukocytes into the bloodstream. Our study illustrates the value of the little skate as a model organism, particularly in studies of hematopoiesis and potentially for preclinical research on hematological and vascular disorders.


Subject(s)
Chemokine CXCL12/metabolism , Fish Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Leukocytes/metabolism , Leukopoiesis , Receptors, CXCR4/metabolism , Skates, Fish/metabolism , Animals , Benzylamines , Chemokine CXCL12/genetics , Cyclams , Fish Proteins/genetics , Gene Expression Regulation , Hematopoietic Stem Cells/drug effects , Heterocyclic Compounds/pharmacology , Leukocytes/drug effects , Leukopoiesis/drug effects , Leukopoiesis/genetics , Phylogeny , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Signal Transduction , Skates, Fish/genetics , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL