Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Blood Cells Mol Dis ; 85: 102485, 2020 11.
Article in English | MEDLINE | ID: mdl-32836190

ABSTRACT

In vitro growth of hematopoietic cells depends on the presence of hematopoietic cytokines. To date, it is unclear if these cells would be able to respond to non-hematopoietic cytokines. In the present study, we have explored this by culturing human hematopoietic cells in presence of neurogenic cytokines. Lineage-negative (Lin-) umbilical cord blood (UCB)-derived cells -enriched for hematopoietic stem and progenitor cells- were cultured in presence of different combinations of hematopoietic cytokines, neurotrophins, epidermal growth factor, fibroblast growth factor, and neurogenic culture media, in a 3-phase culture system. A proportion (1-22%) of Lin- UCB hematopoietic cells normally express neural markers and are capable of responding to neural cytokines. Neural cytokines did not have effects on hematopoietic cell proliferation; however, we observed generation of neural-like cells, assessed by morphology, and a significant increase in the proportion of cells expressing neural markers. Such neural-like cells, however, retained expression of hematopoietic markers. It seems that under our culture conditions, no actual transdifferentiation of hematopoietic cells into neural cells occurred; instead, the cells generated in culture seem to be hematopoietic cells that acquired neural features upon contact with neurogenic factors. The identity of UCB cells that acquired a neural phenotype is still unclear.


Subject(s)
Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Neurogenesis , Cell Culture Techniques , Cells, Cultured , Cytokines/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Nerve Growth Factors/metabolism , Neurons/cytology , Neurons/metabolism
2.
Stem Cell Reports ; 19(1): 100-111, 2024 01 09.
Article in English | MEDLINE | ID: mdl-38101400

ABSTRACT

Lymphoid-primed multipotent progenitor (LMPP)-like and granulocyte-monocyte progenitor (GMP)-like leukemia stem cells (LSCs) co-exist in the blood of most patients with acute myeloid leukemia (AML). Complete elimination of both types of LSCs is required to cure AML. Using an MLL-AF9-induced murine AML model, we studied the role of hematopoietic cytokines in the survival of LMPP- and GMP-like LSCs. We found that SCF or FLT3L promotes the survival of LMPP-like LSCs by stimulating Stat5-mediated Mcl1 expression, whereas interleukin-3 (IL-3) or IL-6 induces the survival of GMP-like LSCs by stimulating Stat3/nuclear factor κB (NF-κB)-mediated Bcl2 expression. Functional study demonstrated that, compared to AML cells cultured in IL-3 and IL-6 medium, AML cells in SCF- or Flt3L-only culture are highly clonogenic in in vitro culture and are highly leukemogenic in vivo. Our study suggests that co-inhibition of both STAT5-MCL1 and STAT3/NF-κB-BCL2 signaling might represent an improved treatment strategy against AML, specifically AML cases with a monocytic phenotype and/or FLT3 mutations.


Subject(s)
Interleukin-3 , Leukemia, Myeloid, Acute , Mice , Humans , Animals , Interleukin-3/metabolism , STAT5 Transcription Factor/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , NF-kappa B/metabolism , Interleukin-6/metabolism , Leukemia, Myeloid, Acute/genetics , Hematopoietic Stem Cells/metabolism , Neoplastic Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism
3.
Front Immunol ; 13: 813676, 2022.
Article in English | MEDLINE | ID: mdl-35250989

ABSTRACT

Polygonatum sibiricum Red. has been used as a medicinal herb and nutritional food in traditional Chinese medicine for a long time. It must be processed prior to clinical use for safe and effective applications. However, the present studies mainly focused on crude Polygonatum sibiricum (PS). This study aimed to investigate the chemical properties, blood-enriching effects and mechanism of polysaccharide from the steam-processed Polygonatum sibiricum (SPS), which is a common form of PS in clinical applications. Instrumentation analyses and chemistry analyses revealed the structure of SPS polysaccharide (SPSP). A mice model of blood deficiency syndrome (BDS) was induced by acetylphenylhydrazine (APH) and cyclophosphamide (CTX). Blood routine test, spleen histopathological changes, serum cytokines, etc. were measured. The spleen transcriptome changes of BDS mice were detected by RNA sequencing (RNA-seq). The results showed that SPSP consists predominantly of Gal and GalA together with fewer amounts of Man, Glc, Ara, Rha and GlcN. It could significantly increase peripheral blood cells, restore the splenic trabecular structure, and reverse hematopoietic cytokines to normal levels. RNA-seq analysis showed that 122 differentially expressed genes (DEGs) were obtained after SPSP treatment. GO and KEGG analysis revealed that SPSP-regulated DEGs were mainly involved in hematopoiesis, immune regulation signaling pathways. The reliability of transcriptome profiling was validated by quantitative real-time PCR and Western blot, and the results indicated that the potential molecular mechanisms of the blood-enriching effects of SPSP might be associated with the regulating of JAK1-STAT1 pathway, and elevated the hematopoietic cytokines (EPO, G-CSF, TNF-α and IL-6). This work provides important information on the potential mechanisms of SPSP against BDS.


Subject(s)
Hematologic Diseases , Polygonatum , Polysaccharides , Animals , Cytokines/metabolism , Hematologic Diseases/immunology , Hematologic Diseases/metabolism , Mice , Polygonatum/chemistry , Polygonatum/metabolism , Polysaccharides/metabolism , Polysaccharides/pharmacology , Reproducibility of Results , Steam
4.
Cannabis Cannabinoid Res ; 7(4): 415-423, 2022 08.
Article in English | MEDLINE | ID: mdl-33998887

ABSTRACT

Introduction: The hematopoietic cytokine granulocyte-colony stimulating factor (G-CSF) is well known to stimulate proliferation of blood stem/progenitor cells of the leukocyte lineage, but is also recognized as a neurotrophic factor involved in brain self-repair processes. G-CSF administration has been shown to promote recovery from experimental models of traumatic brain injury (TBI) and to modulate components of the endocannabinoid system (eCS). Conversely, Δ9-tetrahydrocannabinol (Δ9THC) treatment of normal mice has been shown to increase blood levels of G-CSF in the periphery. Hypothesis: Administration of the phytocannabinoid Δ9THC will enhance brain repair following controlled cortical impact (CCI) by upregulating G-CSF and other neurotrophic factors (brain-derived neurotrophic factor [BDNF] and glial-derived neurotrophic factor [GDNF]) in brain regions. Materials and Methods: C57BL/6J mice underwent CCI and were treated for 3 days with THC 3 mg/kg intraperitoneally. Motor function on a rotarod was recorded at baseline and 3, 7, and 14 days after CCI. Groups of mice were euthanized at 7 and 14 days. G-CSF, BDNF, and GDNF expression were measured at 7 and 14 days in cerebral cortex, striatum, and hippocampus on the side of the trauma. Results: Δ9THC-treated mice ran on the rotarod longer than vehicle-treated mice and recovered to normal rotarod performance levels at 2 weeks. These mice, compared to vehicle-treated animals, exhibited significant upregulation of G-CSF as well as BDNF and GDNF in cerebral cortex, striatum, and hippocampus. Conclusion: Administration of the phytocannabinoid Δ9THC promotes significant recovery from TBI and is associated with upregulation of brain G-CSF, BDNF, and GDNF, neurotrophic factors previously shown to mediate brain self-repair following TBI and stroke.


Subject(s)
Brain Injuries, Traumatic , Brain-Derived Neurotrophic Factor , Animals , Brain Injuries, Traumatic/drug therapy , Disease Models, Animal , Dronabinol/pharmacology , Glial Cell Line-Derived Neurotrophic Factor , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocytes/metabolism , Mice , Mice, Inbred C57BL , Recovery of Function/physiology
5.
Data Brief ; 11: 277-283, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28275660

ABSTRACT

GM-CSF and G-CSF are widely used for their benefit in reducing chemotherapy-associated neutropenia. However, whether GM- or G-CSF administration could have tumorigenic or pro-metastatic effects or whether insulin resistance could negatively impact such effects is not known. Their ability to stimulate monocyte production at the same time with the highly sought after neutrophils' production, enables an enhanced potential for activation of tumor-associated macrophages. At the same time, IL-7 remains the main driver of B and T cell differentiation and maturation, a process linked to the development of insulin resistance and response to diabetes pharmacotherapy. Insulin secretagogues have the potential to interfere with the hematopoiesis process, respectively with the formation of lineages that may lead to a tumorigenic or pro-metastatic phenotype, but this relationship has not been yet investigated. The data presented here shows the relationship between pre-existing use of insulin secretagogues in women diagnosed with breast cancer and type 2 diabetes mellitus, the GM-CSF, G-CSF and IL-7 cytokine profiles at the time of breast cancer diagnosis, and subsequent cancer outcomes. A Pearson correlation analysis evaluating the relationship between investigated cytokines stratified by secretagogue use and controls, and interferon is also provided.

6.
Data Brief ; 11: 382-390, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28275672

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) and granulocyte macrophage colony-stimulating factor (GM-CSF) are cytokines of particular interest in oncology from the perspective of neutropenia management (Mehta et al., 2015 [1]) and also as indirect activators of tumor-associated macrophages and modifiers of tumor microenvironment. Associated with poor breast cancer survival and unfavorable hormone receptor status (Wintrob et al., 2017 [2]), insulin may also influence hematopoiesis, thus interfering with colony stimulating factor production. Although G-CSF has been linked to exacerbating insulin resistance (Ordelheide et al., 2016 [3]), thus far no study linked insulin treatment and hematopoietic cytokines production. Additionally, IL-7 is the primary driver of T and B cell differentiation, maturation, and response (Corfe and Paige, 2012 [4]) and its elevated levels have been associated with poor prognosis in breast cancer. The data presented here is among the first to show a relationship between pre-existing use of injectable insulin in women diagnosed with breast cancer and type 2 diabetes mellitus, hematopoietic cytokine profiles at time of breast cancer diagnosis, and subsequent cancer outcomes. A Pearson correlation analysis evaluating the relationship between G-CSF, GM-CSF, and IL-7 stratified by insulin use, controls, as well as by estrogen and progesterone receptor status is also provided.

7.
J Am Coll Cardiol ; 63(10): 1011-9, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24184252

ABSTRACT

OBJECTIVES: The goal of this study was to define the role of FMS-like tyrosine kinase 3 (FLT3) in the heart. BACKGROUND: FLT3 is a prominent target of receptor tyrosine kinase inhibitors (TKIs) used for anticancer therapy. TKIs can cause cardiomyopathy but understanding of the mechanisms is incomplete, partly because the roles of specific TKI target receptors in the heart are still obscure. METHODS: Myocardial infarction was induced in mice by permanent ligation of the left anterior descending coronary artery followed by intramyocardial injection of FLT3 ligand (FL) or vehicle into the infarct border zone. Cardiac morphology and function were assessed by echocardiography and histological analysis 1 week after infarction. In addition, FLT3 expression and regulation, as well as molecular mechanisms of FLT3 action, were examined in cardiomyocytes in vitro. RESULTS: The intramyocardial injection of FL into the infarct border zone decreased infarct size and ameliorated post-myocardial infarction remodeling and function in mice. This beneficial effect was associated with reduced apoptosis, including myocytes in the infarct border zone. Cardiomyocytes expressed functional FLT3, and FLT3 messenger ribonucleic acid and protein were up-regulated under oxidative stress, identifying cardiomyocytes as FL target cells. FLT3 activation with FL protected cardiomyocytes from oxidative stress-induced apoptosis via an Akt-dependent mechanism involving Bcl-2 family protein regulation and inhibition of the mitochondrial death pathway. CONCLUSIONS: FLT3 is a cytoprotective system in the heart and a potential therapeutic target in ischemic cardiac injury. The protective mechanisms uncovered here may be further explored in view of potential cardiotoxic effects of FLT3-targeting anticancer therapy, particularly in patients with ischemic heart disease.


Subject(s)
Membrane Proteins/administration & dosage , Myocardial Infarction/drug therapy , Myocytes, Cardiac/drug effects , Ventricular Remodeling/physiology , Adjuvants, Immunologic/administration & dosage , Animals , Apoptosis , Cells, Cultured , Disease Models, Animal , Female , Injections, Intralesional , Ligands , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley
9.
Biologics ; 2(1): 13-27, 2008 Mar.
Article in English | MEDLINE | ID: mdl-19707424

ABSTRACT

The role of granulocyte-macrophage-colony-stimulating factor (GM-CSF) in the supportive care of cancer patients has been evaluated with promising results. More recently, GM-CSF has been added to regimens for the mobilization of hematopoietic progenitor cells. An expanding role for GM-CSF in regulating immune responses has been recognized based upon its activity on the development and maturation of antigen presenting cells and its capability for skewing the immune system toward Th1-type responses. GM-CSF has been shown to preferentially enhance both the numbers and activity of type 1 dendritic cells (DC1), the subsets of dendritic cells responsible for initiating cytotoxic immune responses. The increase in DC1 content and activity following local and systemic GM-CSF administration support a role for GM-CSF as an immune stimulant and vaccine adjuvant in cancer patients. GM-CSF has shown clinical activity as an immune stimulant in tumor cell and dendritic cell vaccines, and may increase antibody-dependent cellular cytotoxicity. The successful use of myeloid acting cytokines to enhance anti-tumor responses will likely require the utilization of GM-CSF in combination with cytotoxic or other targeted therapies.

SELECTION OF CITATIONS
SEARCH DETAIL