Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.400
Filter
1.
Front Immunol ; 15: 1361596, 2024.
Article in English | MEDLINE | ID: mdl-38690266

ABSTRACT

Mesenchymal stromal/stem cells (MSCs), which are distributed in many tissues including bone marrow, have been reported to play a critical role in tumor development. While bone marrow, the primary site for hematopoiesis, is important for establishing the immune system, whether MSCs in the bone marrow can promote tumor growth via influencing hematopoiesis remains unclear. We observed that the numbers of MSCs and neutrophils were increased in bone marrow in tumor-bearing mice. Moreover, co-culture assay showed that MSCs strongly protected neutrophils from apoptosis and induced their maturation. G-CSF and GM-CSF have been well-documented to be associated with neutrophil formation. We found a remarkably increased level of G-CSF, but not GM-CSF, in the supernatant of MSCs and the serum of tumor-bearing mice. The G-CSF expression can be enhanced with inflammatory cytokines (IFNγ and TNFα) stimulation. Furthermore, we found that IFNγ and TNFα-treated MSCs enhanced their capability of promoting neutrophil survival and maturation. Our results indicate that MSCs display robustly protective effects on neutrophils to contribute to tumor growth in bone niches.


Subject(s)
Cytokines , Mesenchymal Stem Cells , Neutrophils , Animals , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Mice , Cytokines/metabolism , Mice, Inbred C57BL , Coculture Techniques , Granulocyte Colony-Stimulating Factor/metabolism , Apoptosis , Tumor Necrosis Factor-alpha/metabolism , Cell Line, Tumor , Neoplasms/immunology , Neoplasms/pathology
2.
Bull Exp Biol Med ; 176(5): 607-611, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38730105

ABSTRACT

The study presents the killer functions of circulating neutrophils: myeloperoxidase activity, the ability to generate ROS, phagocytic activity, receptor status, NETosis, as well as the level of cytokines IL-2, IL-4, IL-6, IL-17A, and IL-18, granulocyte CSF, monocyte chemotactic protein 1, and neutrophil elastase in the serum of patients with uterine myoma and endometrial cancer (FIGO stages I-III). The phagocytic ability of neutrophils in uterine myoma was influenced by serum levels of granulocyte CSF and IL-2 in 54% of the total variance. The degranulation ability of neutrophils in endometrial cancer was determined by circulating IL-18 in 50% of the total variance. In uterine myoma, 66% of the total variance in neutrophil myeloperoxidase activity was explained by a model dependent on blood levels of IL-17A, IL-6, and IL-4. The risk of endometrial cancer increases when elevated levels of monocyte chemotactic protein 1 in circulating neutrophils are associated with reduced ability to capture particles via extracellular traps (96% probability).


Subject(s)
Chemokine CCL2 , Endometrial Neoplasms , Interleukin-17 , Interleukin-6 , Neutrophils , Humans , Female , Neutrophils/metabolism , Neutrophils/immunology , Endometrial Neoplasms/immunology , Endometrial Neoplasms/blood , Endometrial Neoplasms/pathology , Endometrial Neoplasms/metabolism , Interleukin-6/blood , Chemokine CCL2/blood , Interleukin-17/blood , Middle Aged , Interleukin-4/blood , Peroxidase/blood , Peroxidase/metabolism , Interleukin-18/blood , Uterine Neoplasms/blood , Uterine Neoplasms/immunology , Uterine Neoplasms/pathology , Granulocyte Colony-Stimulating Factor/blood , Granulocyte Colony-Stimulating Factor/metabolism , Phagocytosis , Leiomyoma/blood , Leiomyoma/immunology , Leiomyoma/pathology , Leiomyoma/metabolism , Cytokines/blood , Cytokines/metabolism , Leukocyte Elastase/blood , Leukocyte Elastase/metabolism , Adult , Extracellular Traps/metabolism , Extracellular Traps/immunology , Reactive Oxygen Species/metabolism , Aged , Interleukin-2
3.
Int J Mol Sci ; 25(10)2024 May 14.
Article in English | MEDLINE | ID: mdl-38791393

ABSTRACT

Lipopolysaccharide-induced (LPS) inflammation is used as model to understand the role of inflammation in brain diseases. However, no studies have assessed the ability of peripheral low-level chronic LPS to induce neutrophil activation in the periphery and brain. Subclinical levels of LPS were injected intraperitoneally into mice to investigate its impacts on neutrophil frequency and activation. Neutrophil activation, as measured by CD11b expression, was higher in LPS-injected mice compared to saline-injected mice after 4 weeks but not 8 weeks of injections. Neutrophil frequency and activation increased in the periphery 4-12 h and 4-8 h after the fourth and final injection, respectively. Increased levels of G-CSF, TNFa, IL-6, and CXCL2 were observed in the plasma along with increased neutrophil elastase, a marker of neutrophil extracellular traps, peaking 4 h following the final injection. Neutrophil activation was increased in the brain of LPS-injected mice when compared to saline-injected mice 4-8 h after the final injection. These results indicate that subclinical levels of peripheral LPS induces neutrophil activation in the periphery and brain. This model of chronic low-level systemic inflammation could be used to understand how neutrophils may act as mediators of the periphery-brain axis of inflammation with age and/or in mouse models of neurodegenerative or neuroinflammatory disease.


Subject(s)
Brain , Lipopolysaccharides , Neutrophil Activation , Neutrophils , Animals , Mice , Brain/metabolism , Brain/drug effects , Neutrophils/metabolism , Neutrophils/immunology , Pilot Projects , Disease Models, Animal , Male , Inflammation/metabolism , Inflammation/chemically induced , Mice, Inbred C57BL , Granulocyte Colony-Stimulating Factor/metabolism , Leukocyte Elastase/metabolism
4.
Nat Immunol ; 25(6): 957-968, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38811815

ABSTRACT

The adult central nervous system (CNS) possesses a limited capacity for self-repair. Severed CNS axons typically fail to regrow. There is an unmet need for treatments designed to enhance neuronal viability, facilitate axon regeneration and ultimately restore lost neurological functions to individuals affected by traumatic CNS injury, multiple sclerosis, stroke and other neurological disorders. Here we demonstrate that both mouse and human bone marrow neutrophils, when polarized with a combination of recombinant interleukin-4 (IL-4) and granulocyte colony-stimulating factor (G-CSF), upregulate alternative activation markers and produce an array of growth factors, thereby gaining the capacity to promote neurite outgrowth. Moreover, adoptive transfer of IL-4/G-CSF-polarized bone marrow neutrophils into experimental models of CNS injury triggered substantial axon regeneration within the optic nerve and spinal cord. These findings have far-reaching implications for the future development of autologous myeloid cell-based therapies that may bring us closer to effective solutions for reversing CNS damage.


Subject(s)
Axons , Granulocyte Colony-Stimulating Factor , Interleukin-4 , Mice, Inbred C57BL , Nerve Regeneration , Neutrophils , Animals , Neutrophils/immunology , Nerve Regeneration/immunology , Mice , Humans , Axons/metabolism , Axons/physiology , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Interleukin-4/metabolism , Neutrophil Activation , Spinal Cord Injuries/therapy , Spinal Cord Injuries/immunology , Spinal Cord Injuries/metabolism , Adoptive Transfer , Cytokines/metabolism , Cells, Cultured
5.
Int Immunopharmacol ; 133: 112058, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38613883

ABSTRACT

Fetal growth restriction (FGR) is a major cause of premature and low-weight births, which increases the risk of necrotizing enterocolitis (NEC); however, the association remains unclear. We report a close correlation between placental polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) and NEC. Newborns with previous FGR exhibited intestinal inflammation and more severe NEC symptoms than healthy newborns. Placental PMN-MDSCs are vital regulators of fetal development and neonatal gut inflammation. Placental single-cell transcriptomics revealed that PMN-MDSCs populations and olfactomedin-4 gene (Olfm4) expression levels were significantly increased in PMN-MDSCs in later pregnancy compared to those in early pregnancy and non-pregnant females. Female mice lacking Olfm4 in myeloid cells mated with wild-type males showed FGR during pregnancy, with a decreased placental PMN-MDSCs population and expression of growth-promoting factors (GPFs) from placental PMN-MDSCs. Galectin-3 (Gal-3) stimulated the OLFM4-mediated secretion of GPFs by placental PMN-MDSCs. Moreover, GPF regulation via OLFM4 in placental PMN-MDSCs was mediated via hypoxia inducible factor-1α (HIF-1α). Notably, the offspring of mothers lacking Olfm4 exhibited intestinal inflammation and were susceptible to NEC. Additionally, OLFM4 expression decreased in placental PMN-MDSCs from pregnancies with FGR and was negatively correlated with neonatal morbidity. These results revealed that placental PMN-MDSCs contributed to fetal development and ameliorate newborn intestinal inflammation.


Subject(s)
Fetal Growth Retardation , Myeloid-Derived Suppressor Cells , Placenta , Animals , Female , Pregnancy , Humans , Placenta/immunology , Placenta/metabolism , Infant, Newborn , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Fetal Growth Retardation/immunology , Mice , Mice, Knockout , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/genetics , Mice, Inbred C57BL , Male , Galectins/metabolism , Galectins/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intestines/immunology , Intestines/pathology
6.
J Innate Immun ; 16(1): 262-282, 2024.
Article in English | MEDLINE | ID: mdl-38643762

ABSTRACT

INTRODUCTION: Signal transducer and activator of transcription (STAT) 3 is extensively involved in the development, homeostasis, and function of immune cells, with STAT3 disruption associated with human immune-related disorders. The roles ascribed to STAT3 have been assumed to be due to its canonical mode of action as an inducible transcription factor downstream of multiple cytokines, although alternative noncanonical functional modalities have also been identified. The relative involvement of each mode was further explored in relevant zebrafish models. METHODS: Genome editing with CRISPR/Cas9 was used to generate mutants of the conserved zebrafish Stat3 protein: a loss of function knockout (KO) mutant and a mutant lacking C-terminal sequences including the transactivation domain (ΔTAD). Lines harboring these mutations were analyzed with respect to blood and immune cell development and function in comparison to wild-type zebrafish. RESULTS: The Stat3 KO mutant showed perturbation of hematopoietic lineages throughout primitive and early definitive hematopoiesis. Neutrophil numbers did not increase in response to lipopolysaccharide (LPS) or granulocyte colony-stimulating factor (G-CSF) and their migration was significantly diminished, the latter correlating with abrogation of the Cxcl8b/Cxcr2 pathway, with macrophage responses perturbed. Intriguingly, many of these phenotypes were not shared by the Stat3 ΔTAD mutant. Indeed, only neutrophil and macrophage development were disrupted in these mutants with responsiveness to LPS and G-CSF maintained, and neutrophil migration actually increased. CONCLUSION: This study has identified roles for zebrafish Stat3 within hematopoietic stem cells impacting multiple lineages throughout primitive and early definitive hematopoiesis, myeloid cell responses to G-CSF and LPS and neutrophil migration. Many of these roles showed conservation, but notably several involved noncanonical modalities, providing additional insights for relevant diseases.


Subject(s)
Hematopoiesis , STAT3 Transcription Factor , Zebrafish Proteins , Zebrafish , Animals , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Hematopoiesis/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Humans , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neutrophils/immunology , Signal Transduction , CRISPR-Cas Systems , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/genetics , Gene Editing , Lipopolysaccharides , Hematopoietic Stem Cells
7.
Cytokine ; 179: 156619, 2024 07.
Article in English | MEDLINE | ID: mdl-38669908

ABSTRACT

Interleukin (IL)-23 is implicated in the pathogenesis of several inflammatory diseases and is usually linked with helper T cell (Th17) biology. However, there is some data linking IL-23 with innate immune biology in such diseases. We therefore examined the effects of IL-23p19 genetic deletion and/or neutralization on in vitro macrophage activation and in an innate immune-driven peritonitis model. We report that endogenous IL-23 was required for maximal macrophage activation by zymosan as determined by pro-inflammatory cytokine production, including a dramatic upregulation of granulocyte-colony stimulating factor (G-CSF). Furthermore, both IL-23p19 genetic deletion and neutralization in zymosan-induced peritonitis (ZIP) led to a specific reduction in the neutrophil numbers, as well as a reduction in the G-CSF levels in exudate fluids. We conclude that endogenous IL-23 can contribute significantly to macrophage activation during an inflammatory response, mostly likely via an autocrine/paracrine mechanism; of note, endogenous IL-23 can directly up-regulate macrophage G-CSF expression, which in turn is likely to contribute to the regulation of IL-23-dependent neutrophil number and function during an inflammatory response, with potential significance for IL-23 targeting particularly in neutrophil-associated inflammatory diseases.


Subject(s)
Inflammation , Interleukin-23 , Myeloid Cells , Neutrophils , Zymosan , Animals , Inflammation/metabolism , Inflammation/immunology , Interleukin-23/metabolism , Mice , Neutrophils/metabolism , Neutrophils/immunology , Myeloid Cells/metabolism , Peritonitis/metabolism , Peritonitis/immunology , Mice, Inbred C57BL , Granulocyte Colony-Stimulating Factor/metabolism , Macrophage Activation , Macrophages/metabolism , Macrophages/immunology , Interleukin-23 Subunit p19/metabolism , Interleukin-23 Subunit p19/genetics , Mice, Knockout
8.
Sci Rep ; 14(1): 7292, 2024 03 27.
Article in English | MEDLINE | ID: mdl-38538691

ABSTRACT

Glioblastoma multiforme (GBM) IDH-wildtype is the most prevalent brain malignancy in adults. However, molecular mechanisms, which leads to GBM have not been completely elucidated. Granulocyte colony-stimulating factor (GCSF), Granulocyte colony-stimulating factor receptor GCSFR, and Signal transducers and activators of transcription 3 (STAT3) have been involved in the occurrence and development of various cancers, but their role in GBM is little known. Herein, we have investigated the gene and protein expression of GCSF, GCSFR, and STAT3 in 21 tissue biopsy samples and also in tumor associated normal tissue (TANT) samples derived from glioblastoma patients, which revealed significantly differential expression of these genes. To validate our findings, we performed a comprehensive integrated analysis of transcriptomic and proteomic profiling of respective genes by retrieving GBM RNA-sequence data from Genome Atlas Databases. GO and KEGG analysis revealed enrichment in disease-related pathways, such as JAK/STAT pathway activation, which were associated with GBM progression. We further performed computational docking analysis of potential drug candidate Nisin against GCSF, and the results were validated in vitro through cytotoxic activity assay using a human glioblastoma cell line SF-767 in a dose-dependent manner. Our comprehensive analysis reveals that GCSF augments glioma progression, and its blockade with anticancer bacteriocin peptide Nisin can potentially inhibit the growth and metastasis of GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Nisin , Adult , Humans , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Nisin/metabolism , Janus Kinases/metabolism , Proteomics , Signal Transduction , STAT Transcription Factors/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Granulocyte Colony-Stimulating Factor/metabolism , Gene Expression Regulation, Neoplastic
9.
Environ Int ; 186: 108580, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38507932

ABSTRACT

Desert dust exposure is associated with adverse respiratory health effects. Desert dust is a complex pollutant mixtures that includes respirable crystalline and amorphous particles, metals, and microbial constituents. Given the health effects of desert dust and its heterogeneity, as yet unidentified harmful biological pathways may be triggered. Therefore, we exposed human in vitro air-liquid interface co-cultures of alveolar epithelial A549 cells and THP-1 macrophages to Saharan dust (SD). For comparison, we used the known pulmonary toxicant DQ12 quartz dust. Via RNA sequencing, we identified that SD but not DQ12 increased the gene expression of granulocyte-macrophage colony-stimulating factor (GMCSF) and granulocyte colony-stimulating factor (GCSF). These findings were confirmed by quantitative reverse transcriptase PCR. SD dose-dependently upregulated GMCSF and GCSF expression with significant 7 and 9-fold changes, respectively, at the highest tested concentration of 31 µg/cm2. Furthermore, we observed that SD significantly enhanced the secretion of GM-CSF and G-CSF by 2-fold. Both cytokines have previously been associated with lung diseases such as asthma and fibrosis. Hence, we present two molecular messengers that may contribute to the adverse health effects of desert dust and might serve as drug targets for this globally relevant non-anthropogenic air pollutant.


Subject(s)
Dust , Granulocyte Colony-Stimulating Factor , Granulocyte-Macrophage Colony-Stimulating Factor , Lung Diseases , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Granulocyte Colony-Stimulating Factor/metabolism , Lung Diseases/chemically induced , A549 Cells , THP-1 Cells , Cytokines/metabolism
10.
Nature ; 627(8005): 839-846, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38509363

ABSTRACT

The bone marrow adjusts blood cell production to meet physiological demands in response to insults. The spatial organization of normal and stress responses are unknown owing to the lack of methods to visualize most steps of blood production. Here we develop strategies to image multipotent haematopoiesis, erythropoiesis and lymphopoiesis in mice. We combine these with imaging of myelopoiesis1 to define the anatomy of normal and stress haematopoiesis. In the steady state, across the skeleton, single stem cells and multipotent progenitors distribute through the marrow enriched near megakaryocytes. Lineage-committed progenitors are recruited to blood vessels, where they contribute to lineage-specific microanatomical structures composed of progenitors and immature cells, which function as the production sites for each major blood lineage. This overall anatomy is resilient to insults, as it was maintained after haemorrhage, systemic bacterial infection and granulocyte colony-stimulating factor (G-CSF) treatment, and during ageing. Production sites enable haematopoietic plasticity as they differentially and selectively modulate their numbers and output in response to insults. We found that stress responses are variable across the skeleton: the tibia and the sternum respond in opposite ways to G-CSF, and the skull does not increase erythropoiesis after haemorrhage. Our studies enable in situ analyses of haematopoiesis, define the anatomy of normal and stress responses, identify discrete microanatomical production sites that confer plasticity to haematopoiesis, and uncover unprecedented heterogeneity of stress responses across the skeleton.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells , Stress, Physiological , Animals , Female , Male , Mice , Aging/physiology , Bacterial Infections/pathology , Bacterial Infections/physiopathology , Blood Vessels/cytology , Cell Lineage , Erythropoiesis , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hemorrhage/pathology , Hemorrhage/physiopathology , Lymphopoiesis , Megakaryocytes/cytology , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Myelopoiesis , Skull/blood supply , Skull/pathology , Skull/physiopathology , Sternum/blood supply , Sternum/cytology , Sternum/metabolism , Stress, Physiological/physiology , Tibia/blood supply , Tibia/cytology , Tibia/metabolism
11.
Biochem Biophys Res Commun ; 704: 149661, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38417343

ABSTRACT

To date only four recombinant growth factors, including Filgrastim (rhG-CSF), have been approved by FDA as radiomitigators to ameliorate hematopoietic acute radiation syndrome (H-ARS). These approved agents are not stable under room-temperature, needing to be stored at 2-8 °C, and would not be feasible in a mass casualty scenario where rapid and cost-effective intervention is crucial. Delta-tocotrienol (δ-T3H), the most potent G-CSF-inducing agent among vitamin E isoforms, exhibited efficiency and selectivity on G-CSF production in comparison with TLR and STING agonists in mice. Five-dose δ-T3H was utilized as the optimal therapeutic regimen due to long-term G-CSF production and the best peripheral blood (PB) recovery of irradiated mice. Comparable with rhG-CSF, sequential administration of δ-T3H post-irradiation improved hematologic recovery and accelerated the regeneration of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) in the bone marrow (BM) and spleen of 6.5Gy irradiated mice; and consistently enhanced repopulation of BM-HSCs. In 4.0Gy irradiated nonhuman primates, δ-T3H exhibited comparable efficacy as rhG-CSF to promote PB recovery and colony-formation of BM-HPCs. Altogether, we demonstrated that sequential administration of delta-tocotrienol ameliorates radiation-induced myelosuppression in mice and non-human primates through inducing G-CSF production, indicated δ-T3H as a promising radiomitigator for the management of H-ARS, particularly in a mass casualty scenario.


Subject(s)
Bone Marrow , Hematopoietic Stem Cells , Vitamin E , Animals , Mice , Bone Marrow/pathology , Bone Marrow/radiation effects , Granulocyte Colony-Stimulating Factor/drug effects , Granulocyte Colony-Stimulating Factor/metabolism , Primates , Recombinant Proteins/pharmacology , Vitamin E/analogs & derivatives , Vitamin E/therapeutic use
12.
Proc Natl Acad Sci U S A ; 121(6): e2305947121, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38289952

ABSTRACT

Optic neuropathies, characterized by injury of retinal ganglion cell (RGC) axons of the optic nerve, cause incurable blindness worldwide. Mesenchymal stem cell-derived small extracellular vesicles (MSC-sEVs) represent a promising "cell-free" therapy for regenerative medicine; however, the therapeutic effect on neural restoration fluctuates, and the underlying mechanism is poorly understood. Here, we illustrated that intraocular administration of MSC-sEVs promoted both RGC survival and axon regeneration in an optic nerve crush mouse model. Mechanistically, MSC-sEVs primarily targeted retinal mural cells to release high levels of colony-stimulating factor 3 (G-CSF) that recruited a neural restorative population of Ly6Clow monocytes/monocyte-derived macrophages (Mo/MΦ). Intravitreal administration of G-CSF, a clinically proven agent for treating neutropenia, or donor Ly6Clow Mo/MΦ markedly improved neurological outcomes in vivo. Together, our data define a unique mechanism of MSC-sEV-induced G-CSF-to-Ly6Clow Mo/MΦ signaling in repairing optic nerve injury and highlight local delivery of MSC-sEVs, G-CSF, and Ly6Clow Mo/MΦ as therapeutic paradigms for the treatment of optic neuropathies.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Optic Nerve Injuries , Mice , Animals , Axons/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Nerve Regeneration/physiology , Optic Nerve Injuries/therapy , Optic Nerve Injuries/metabolism , Retinal Ganglion Cells/physiology , Mesenchymal Stem Cells/metabolism , Extracellular Vesicles/metabolism , Macrophages/metabolism
13.
Cell Rep Med ; 5(2): 101380, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38242120

ABSTRACT

Precise molecular characterization of circulating polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) is hampered by their mixed composition of mature and immature cells and lack of specific markers. Here, we focus on mature CD66b+CD10+CD16+CD11b+ PMN-MDSCs (mPMN-MDSCs) from either cancer patients or healthy donors receiving G-CSF for stem cell mobilization (GDs). By RNA sequencing (RNA-seq) experiments, we report the identification of a distinct gene signature shared by the different mPMN-MDSC populations under investigation, also validated in mPMN-MDSCs from GDs and tumor-associated neutrophils (TANs) by single-cell RNA-seq (scRNA-seq) experiments. Analysis of such a gene signature uncovers a specific transcriptional program associated with mPMN-MDSC differentiation and allows us to identify that, in patients with either solid or hematologic tumors and in GDs, CD52, CD84, and prostaglandin E receptor 2 (PTGER2) represent potential mPMN-MDSC-associated markers. Altogether, our findings indicate that mature PMN-MDSCs distinctively undergo specific reprogramming during differentiation and lay the groundwork for selective immunomonitoring, and eventually targeting, of mature PMN-MDSCs.


Subject(s)
Myeloid-Derived Suppressor Cells , Neoplasms , Humans , Neutrophils , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Colony-Stimulating Factor/metabolism , Neoplasms/pathology , CD52 Antigen/metabolism , Signaling Lymphocytic Activation Molecule Family/metabolism
15.
Intern Med ; 63(2): 189-195, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-37225486

ABSTRACT

Objective Several institutions outsource CD34+ cell counting of leukapheresis products, limiting rapid measurements, as results are obtained the next day. This problem is compounded with plerixafor use, a stem cell-mobilizing drug that increases leukapheresis efficiency but requires administration the day before leukapheresis. Use of this drug for a second leukapheresis procedure before the first-day leukapheresis CD34+ count results are confirmed causes unnecessary leukapheresis and expensive plerixafor administration. We investigated whether or not measuring hematopoietic progenitor cells in leukapheresis products (AP-HPCs) using a Sysmex XN-series analyzer could resolve this problem. Methods We retrospectively compared the absolute AP-HPC value per body weight with the CD34+ (AP-CD34+) count in 96 first-day leukapheresis product samples obtained between September 2013 and January 2021. Comparisons were also conducted according to regimen: granulocyte colony-stimulating factor (G-CSF) monotherapy, chemotherapy plus G-CSF, or plerixafor mobilization. Results AP-CD34+ and AP-HPC counts correlated strongly (rs=0.846) overall and, in particular, under chemotherapy plus G-CSF (rs=0.92) but correlated mildly under G-CSF monotherapy (rs=0.655). AP-HPCs could not completely be dichotomized based on an AP-CD34+ threshold of 2×106/kg for any stimulation procedure. In most cases with AP-HPCs >6×106/kg, the AP-CD34+ count exceeded 2.0×106/kg, but in 5.7% of these cases, the AP-CD34+ count was <2.0×106/kg. A cut-off of AP-HPCs >4.843×106/kg yielded a sensitivity of 71% and specificity of 96% for predicting AP-CD34+≥2×106/kg. Conclusion AP-HPCs can identify cases in which sufficient stem cells have been collected.


Subject(s)
Hematopoietic Stem Cell Transplantation , Heterocyclic Compounds , Peripheral Blood Stem Cell Transplantation , Peripheral Blood Stem Cells , Humans , Leukapheresis , Hematopoietic Stem Cell Mobilization/methods , Peripheral Blood Stem Cells/metabolism , Retrospective Studies , Transplantation, Autologous , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/metabolism , Hematopoietic Stem Cells/metabolism , Antigens, CD34/metabolism , Granulocyte Colony-Stimulating Factor/metabolism
16.
Appl Biochem Biotechnol ; 196(2): 804-820, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37209276

ABSTRACT

Granulocyte colony-stimulating factor (GCSF) stimulates the proliferation of neutrophils but it has low serum half-life. Therefore, the present study was done to investigate the effect of XTENylation on biological activity, pharmacokinetics, and pharmacodynamics of GCSF in a neutropenic rat model. XTEN tag was genetically fused to the N-terminal region of GCSF-encoding gene fragment and subcloned into pET28a expression vector. The cytoplasmic expressed recombinant protein was characterized through intrinsic fluorescence spectroscopy (IFS), dynamic light scattering (DLS), and size exclusion chromatography (SEC). In vitro biological activity of the XTEN-GCSF protein was evaluated on NFS60 cell line. Hematopoietic properties and pharmacokinetics were also investigated in a neutropenic rat model. An approximately 140 kDa recombinant protein was detected on SDS-PAGE. Dynamic light scattering and size exclusion chromatography confirmed the increase in hydrodynamic diameter of GCSF molecule after XTENylation. GCSF derivatives showed efficacy in proliferation of NFS60 cell line among which the XTEN-GCSF represented the lowest EC50 value (100.6 pg/ml). Pharmacokinetic studies on neutropenic rats revealed that XTEN polymer could significantly increase protein serum half-life in comparison with the commercially available GCSF molecules. PEGylated and XTENylated GCSF proteins were more effective in stimulation of neutrophils compared to the GCSF molecule alone. XTENylation of GCSF represented promising results in in vitro and in vivo studies. This approach can be a potential alternative to PEGylation strategies for increasing serum half-life of protein.


Subject(s)
Granulocyte Colony-Stimulating Factor , Polymers , Animals , Rats , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte Colony-Stimulating Factor/isolation & purification , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Neutrophils , Polymers/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
17.
Acta Pharmacol Sin ; 45(1): 98-111, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37726422

ABSTRACT

Restenosis after angioplasty is caused usually by neointima formation characterized by aberrant vascular smooth muscle cell (VSMC) dedifferentiation. Myeloid-derived growth factor (MYDGF), secreted from bone marrow-derived monocytes and macrophages, has been found to have cardioprotective effects. In this study we investigated the effect of MYDGF to postinjury neointimal formation and the underlying mechanisms. Rat carotid arteries balloon-injured model was established. We found that plasma MYDGF content and the level of MYDGF in injured arteries were significantly decreased after balloon injury. Local application of exogenous MYDGF (50 µg/mL) around the injured vessel during balloon injury markedly ameliorated the development of neointimal formation evidenced by relieving the narrow endovascular diameter, improving hemodynamics, and reducing collagen deposition. In addition, local application of MYDGF inhibited VSMC dedifferentiation, which was proved by reversing the elevated levels of osteopontin (OPN) protein and decreased levels of α-smooth muscle actin (α-SMA) in the left carotid arteries. We showed that PDGF-BB (30 ng/mL) stimulated VSMC proliferation, migration and dedifferentiation in vitro; pretreatment with MYDGF (50-200 ng/mL) concentration-dependently eliminated PDGF-BB-induced cell proliferation, migration and dedifferentiation. Molecular docking revealed that MYDGF had the potential to bind with sphingosine-1-phosphate receptor 2 (S1PR2), which was confirmed by SPR assay and Co-IP analysis. Pretreatment with CCG-1423 (Rho signaling inhibitor), JTE-013 (S1PR2 antagonist) or Ripasudil (ROCK inhibitor) circumvented the inhibitory effects of MYDGF on VSMC phenotypic switching through inhibiting S1PR2 or its downstream RhoA-actin monomers (G-actin) /actin filaments (F-actin)-MRTF-A signaling. In summary, this study proves that MYDGF relieves neointimal formation of carotid arteries in response to balloon injury in rats, and suppresses VSMC dedifferentiation induced by PDGF-BB via S1PR2-RhoA-G/F-actin-MRTF-A signaling pathway. In addition, our results provide evidence for cross talk between bone marrow and vasculature.


Subject(s)
Actins , Neointima , Rats , Animals , Becaplermin/pharmacology , Neointima/drug therapy , Neointima/metabolism , Actins/metabolism , Rats, Sprague-Dawley , Sphingosine-1-Phosphate Receptors/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Muscle, Smooth, Vascular , Molecular Docking Simulation , Cell Proliferation , Signal Transduction , Cell Movement , Myocytes, Smooth Muscle/metabolism , Cells, Cultured
18.
J Leukoc Biol ; 115(2): 205-221, 2024 01 19.
Article in English | MEDLINE | ID: mdl-37824822

ABSTRACT

Cytosolic proliferating cell nuclear antigen (PCNA) is involved in neutrophil survival and function, in which it acts as a scaffold and associates with proteins involved in apoptosis, NADPH oxidase activation, cytoskeletal dynamics, and metabolism. While the PCNA interactome has been characterized in neutrophils under homeostatic conditions, less is known about neutrophil PCNA in pathophysiological contexts. Granulocyte colony-stimulating factor (G-CSF) is a cytokine produced in response to inflammatory stimuli that regulates many aspects of neutrophil biology. Here, we used isolated normal-density neutrophils from G-CSF-treated haemopoietic stem cell donors (GDs) as a model to understand the role of PCNA during inflammation. Proteomic analysis of the neutrophil cytosol revealed significant differences between GDs and healthy donors (HDs). PCNA was one of the most upregulated proteins in GDs, and the PCNA interactome was significantly different in GDs compared with HDs. Importantly, while PCNA associated with almost all enzymes involved in glycolysis in HDs, these associations were decreased in GDs. Functionally, neutrophils from GDs had a significant increase in glycolysis compared with HDs. Using p21 competitor peptides, we showed that PCNA negatively regulates neutrophil glycolysis in HDs but had no effect on GD neutrophils. These data demonstrate that G-CSF alters the PCNA scaffold, affecting interactions with key glycolytic enzymes, and thus regulates glycolysis, the main energy pathway utilized by neutrophils. By this selective control of glycolysis, PCNA can organize neutrophils functionality in parallel with other PCNA mechanisms of prolonged survival. PCNA may therefore be instrumental in the reprogramming that neutrophils undergo in inflammatory or tumoral settings.


Subject(s)
Granulocyte Colony-Stimulating Factor , Neutrophils , Neutrophils/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Cytosol/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Proteomics , Cytokines/metabolism
19.
Theriogenology ; 216: 93-102, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38159389

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF), a pleiotropic cytokine, is secreted by the reproductive tract. Furthermore, our previous study indicated that human recombinant G-CSF (hrG-CSF) supplementation during porcine oocyte in vitro maturation (IVM) or during embryo in vitro culture (IVC) improved their quality and development potential when using cumulus-oocyte complexes (COCs) with more than three cumulus cell layers (CCL >3). Thus, in this study, we investigate the optimal conditions of hrG-CSF supplementation throughout the in vitro production (IVP: IVM + IVC) system to improve the embryo production efficiency of "poor-quality (CCL ≤3)" oocytes. COCs were classified into two groups according to the number of CCL (>3 and ≤3) and embryonic viability was analyzed after treatment with hrG-CSF during IVC. The mRNA transcription levels of G-CSF in COCs were compared based on their type and the period of IVM. Finally, developmental capacity and quality were evaluated after treatment with hrG-CSF for different periods of IVP. No marked effects on the developmental potential of embryos when using CCL ≤3 type COCs were observed after supplementing hrG-CSF only during IVC. Moreover, the mRNA transcription level of G-CSF increased gradually with IVM culture time and was higher in CCL ≤3 COCs than in >3. Supplementing hrG-CSF only during the IVM period resulted in the best embryo developmental potential, while supplementing hrG-CSF during the IVP period resulted in the best quality embryos, reflected in the increased total cell number and decreased apoptotic nuclei index of blastocysts. These findings indicate that "poor-quality" COCs may have a greater demand for G-CSF than "good-quality", meanwhile hrG-CSF supplementation throughout IVP improves resource utilization efficiency in poor-quality COCs.


Subject(s)
In Vitro Oocyte Maturation Techniques , Oocytes , Female , Humans , Animals , Swine , In Vitro Oocyte Maturation Techniques/veterinary , In Vitro Oocyte Maturation Techniques/methods , Embryonic Development , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Colony-Stimulating Factor/metabolism , Cumulus Cells/metabolism , Blastocyst , RNA, Messenger/metabolism , Dietary Supplements , Granulocytes
20.
Stem Cell Res Ther ; 14(1): 300, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37864264

ABSTRACT

BACKGROUND: Granulocyte colony-stimulating factor (G-CSF)-mediated mobilization of hematopoietic stem cells (HSCs) is a well-established method to prepare HSCs for transplantation nowadays. A sufficient number of HSCs is critical for successful HSC transplantation. However, approximately 2-6% of healthy stem cell donors are G-CSF-poor mobilizers for unknown reasons; thus increasing the uncertainties of HSC transplantation. The mechanism underlining G-CSF-mediated HSC mobilization remains elusive, so detailed mechanisms and an enhanced HSC mobilization strategy are urgently needed. Evidence suggests that P-selectin and P-selectin glycoprotein ligand-1 (PSGL-1) are one of the cell-cell adhesion ligand-receptor pairs for HSCs to keep contacting bone marrow (BM) stromal cells before being mobilized into circulation. This study hypothesized that blockage of PSGL-1 and P-selectin may disrupt HSC-stromal cell interaction and facilitate HSC mobilization. METHODS: The plasma levels of soluble P-selectin (sP-sel) before and after G-CSF administration in humans and male C57BL/6J mice were analyzed using enzyme-linked immunosorbent assay. Male mice with P-selectin deficiency (Selp-/-) were further employed to investigate whether P-selectin is essential for G-CSF-induced HSC mobilization and determine which cell lineage is sP-sel derived from. Finally, wild-type mice were injected with either G-CSF or recombinant sP-sel to investigate whether sP-sel alone is sufficient for inducing HSC mobilization and whether it accomplishes this by binding to HSCs and disrupting their interaction with stromal cells in the BM. RESULTS: A significant increase in plasma sP-sel levels was observed in humans and mice following G-CSF administration. Treatments of G-CSF induced a decrease in the level of HSC mobilization in Selp-/- mice compared with the wild-type (Selp+/+) controls. Additionally, the transfer of platelets derived from wild-type mice can ameliorate the defected HSC mobilization in the Selp-/- recipients. G-CSF induces the release of sP-sel from platelets, which is sufficient to mobilize BM HSCs into the circulation of mice by disrupting the PSGL-1 and P-selectin interaction between HSCs and stromal cells. These results collectively suggested that P-selectin is a critical factor for G-CSF-induced HSC mobilization. CONCLUSIONS: sP-sel was identified as a novel endogenous HSC-mobilizing agent. sP-sel injections achieved a relatively faster and more convenient regimen to mobilize HSCs in mice than G-CSF. These findings may serve as a reference for developing and optimizing human HSC mobilization in the future.


Subject(s)
Hematopoietic Stem Cell Mobilization , P-Selectin , Male , Mice , Humans , Animals , Hematopoietic Stem Cell Mobilization/methods , P-Selectin/genetics , P-Selectin/metabolism , Mice, Inbred C57BL , Hematopoietic Stem Cells/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Colony-Stimulating Factor/metabolism , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...