Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.412
Filter
1.
Mol Cancer ; 23(1): 161, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39118167

ABSTRACT

This commentary offers a thoughtful discussion of the study by Wei et al. published in the journal on the role of Olfactomedin 4 (OLFM4) in incomplete intestinal metaplasia, a gastric precancerous condition. The original paper introduces OLFM4 as a novel biomarker with potential enhanced diagnostic efficacy compared to established markers. However, several methodological and interpretive considerations are noted. The histopathological findings could be refined by using higher magnification to better elucidate the cellular localization of OLFM4. Including high-resolution images for key stainings would enhance the study's robustness in expression profiling. The statistical approach could be strengthened by employing more rigorous, quantitative methodologies. Additionally, integrating immunofluorescence double-staining may improve the reliability of the results. Discrepancies in immunohistochemical signals across datasets suggest a need for further investigation into tissue section representativeness. Clarifying the term "precancerous lesions of gastric carcinoma cells" to align with widely accepted definitions would enhance clarity. The choice of the GES-1 cell model treated with MNNG could be reconsidered in favor of more established models such as organoids, air-liquid interface models, and gastric cancer-specific cell lines. The in vivo MNNG-alcohol combination model might require additional empirical support, given the limited and conflicting literature on this approach, to ensure an accurate portrayal of IM pathogenesis. The commentary concludes with a call for stringent and standardized methodologies in biomarker research to ensure the clinical applicability and reliability of biomarker studies, particularly in the context of gastric cancer detection and intervention.


Subject(s)
Biomarkers, Tumor , Granulocyte Colony-Stimulating Factor , Precancerous Conditions , Stomach Neoplasms , Stomach Neoplasms/diagnosis , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Humans , Precancerous Conditions/diagnosis , Precancerous Conditions/pathology , Granulocyte Colony-Stimulating Factor/metabolism
2.
Immunobiology ; 229(5): 152843, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39186867

ABSTRACT

We previously reported that myeloperoxidase-deficient (MPO-/-) mice develop more severe neutrophil-rich lung inflammation than wild-type mice following intranasal Zymosan administration. Interestingly, we found that these mutant mice with severe lung inflammation also displayed pronounced neutrophilia and anemia, characterized by increased granulopoiesis and decreased erythropoiesis in the bone marrow, compared to wild-type mice. This condition was associated with higher concentrations of granulocyte-colony stimulating factor (G-CSF) in both the lungs and serum, a factor known to enhance granulopoiesis. Neutrophils accumulating in the lungs of MPO-/- mice produced greater amounts of G-CSF than those in wild-type mice, indicating that they are a significant source of G-CSF. In vitro experiments using signal transduction inhibitors and Western blot analysis revealed that MPO-/- neutrophils express higher levels of G-CSF mRNA in response to Zymosan, attributed to the upregulation of the IκB kinase/nuclear factor (NF)-κB pathway and the extracellular-signal-regulated kinase/NF-κB pathway. These findings highlight MPO as a critical regulator of granulopoiesis and erythropoiesis in inflamed tissues.


Subject(s)
Anemia , Erythropoiesis , Granulocyte Colony-Stimulating Factor , Mice, Knockout , Neutrophils , Peroxidase , Pneumonia , Zymosan , Animals , Mice , Neutrophils/immunology , Neutrophils/metabolism , Peroxidase/metabolism , Anemia/etiology , Pneumonia/etiology , Pneumonia/metabolism , Pneumonia/immunology , Granulocyte Colony-Stimulating Factor/metabolism , Signal Transduction , NF-kappa B/metabolism , Granulocytes/metabolism , Granulocytes/immunology , Lung/pathology , Disease Models, Animal , Mice, Inbred C57BL
3.
Int Immunopharmacol ; 141: 112959, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39163688

ABSTRACT

Staphylococcus aureus (S. aureus)-induced bone loss is a significant challenge in the treatment of osteomyelitis. Our previous study was the first to confirm that granulocyte colony-stimulating factor (G-CSF) mediates S. aureus-induced bone loss. However, the underlying mechanism remains unknown. The objective of this study was to elucidate this. We found G-CSF mediated BMSC senescence and increased IL-1ß concentration of serum and bone marrow in mice after S. aureus infection. Furthermore, we demonstrated that G-CSF promoted the expression of IL1b in murine bone marrow-derived neutrophils. Notably, we identified that IL-1ß mediated BMSC (bone marrow mesenchymal stromal cell) senescence in mice after S. aureus infection. Importantly, IL-1ß neutralizing antibody effectively alleviated BMSC senescence and bone loss caused by S. aureus infection in mice. In terms of molecular mechanism, we found IL-1ß induced BMSC senescence by JNK/P53 and JNK/BCL2 pathways. Collectively, G-CSF promotes IL-1ß production which induces BMSC senescence via JNK/P53 and JNK/BCL2 pathways, leading to S. aureus-induced bone loss. This study identified novel targets for preventing and treating S. aureus-induced bone loss in osteomyelitis.


Subject(s)
Granulocyte Colony-Stimulating Factor , Interleukin-1beta , Osteomyelitis , Staphylococcal Infections , Staphylococcus aureus , Animals , Interleukin-1beta/metabolism , Osteomyelitis/microbiology , Osteomyelitis/immunology , Osteomyelitis/metabolism , Staphylococcal Infections/immunology , Granulocyte Colony-Stimulating Factor/metabolism , Mice , Mice, Inbred C57BL , Mesenchymal Stem Cells/metabolism , MAP Kinase Signaling System/drug effects , Neutrophils/immunology , Cellular Senescence/drug effects , Bone Resorption/immunology , Cells, Cultured , Male , Signal Transduction
4.
Biomolecules ; 14(8)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39199390

ABSTRACT

The treatment of patients diagnosed with hematologic malignancies typically includes hematopoietic stem cell transplantation (HSCT) as part of a therapeutic standard of care. The primary graft source of hematopoietic stem and progenitor cells (HSPCs) for HSCT is mobilized from the bone marrow into the peripheral blood of allogeneic donors or patients. More recently, these mobilized HSPCs have also been the source for gene editing strategies to treat diseases such as sickle-cell anemia. For a HSCT to be successful, it requires the infusion of a sufficient number of HSPCs that are capable of adequate homing to the bone marrow niche and the subsequent regeneration of stable trilineage hematopoiesis in a timely manner. Granulocyte-colony-stimulating factor (G-CSF) is currently the most frequently used agent for HSPC mobilization. However, it requires five or more daily infusions to produce an adequate number of HSPCs and the use of G-CSF alone often results in suboptimal stem cell yields in a significant number of patients. Furthermore, there are several undesirable side effects associated with G-CSF, and it is contraindicated for use in sickle-cell anemia patients, where it has been linked to serious vaso-occlusive and thrombotic events. The chemokine receptor CXCR4 and the cell surface integrin α4ß1 (very late antigen 4 (VLA4)) are both involved in the homing and retention of HSPCs within the bone marrow microenvironment. Preclinical and/or clinical studies have shown that targeted disruption of the interaction of the CXCR4 or VLA4 receptors with their endogenous ligands within the bone marrow niche results in the rapid and reversible mobilization of HSPCs into the peripheral circulation and is synergistic when combined with G-CSF. In this review, we discuss the roles CXCR4 and VLA4 play in bone marrow homing and retention and will summarize more recent development of small-molecule CXCR4 and VLA4 inhibitors that, when combined, can synergistically improve the magnitude, quality and convenience of HSPC mobilization for stem cell transplantation and ex vivo gene therapy after the administration of just a single dose. This optimized regimen has the potential to afford a superior alternative to G-CSF for HSPC mobilization.


Subject(s)
Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells , Integrin alpha4beta1 , Receptors, CXCR4 , Humans , Receptors, CXCR4/metabolism , Receptors, CXCR4/antagonists & inhibitors , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/cytology , Integrin alpha4beta1/metabolism , Integrin alpha4beta1/antagonists & inhibitors , Animals , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cell Transplantation
5.
Int J Mol Sci ; 25(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39201584

ABSTRACT

Monocytes are circulating macrophage precursors generated from bone marrow hematopoietic stem cells. In adults, monocytes continuously replenish cerebral border-associated macrophages under physiological conditions. Monocytes also rapidly infiltrate the brain in pathological settings. The mechanisms of recruiting monocyte-derived macrophages into the brain under pathological conditions have been extensively studied. However, it remains unclear how monocytes enter the brain to renew border-associated macrophages under physiological conditions. Using both in vitro and in vivo approaches, this study reveals that a combination of two hematopoietic growth factors, stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF), complementarily and synergistically enhances the adhesion of monocytes to cerebral endothelial cells in a dose-dependent manner. Cysteine-cysteine chemokine receptor 5 (CCR5) in brain endothelial cells, but not the cell adhesion molecules mediating neuroinflammation-related infiltration of monocyte-derived macrophages, modulates SCF+G-CSF-enhanced monocyte-endothelial cell adhesion. Blocking CCR5 or genetically deleting CCR5 reduces monocyte-endothelial cell adhesion induced by SCF+G-CSF. The SCF+G-CSF-enhanced recruitment of bone marrow-derived monocytes/macrophages into the cerebral perivascular space is also reduced in adult CCR5 knockout mice. This study demonstrates the role of SCF and G-CSF in regulating the entry of monocytes into the adult brain to replenish perivascular macrophages.


Subject(s)
Brain , Cell Adhesion , Granulocyte Colony-Stimulating Factor , Monocytes , Receptors, CCR5 , Receptors, CCR5/metabolism , Receptors, CCR5/genetics , Animals , Monocytes/metabolism , Mice , Brain/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Mice, Knockout , Endothelial Cells/metabolism , Stem Cell Factor/metabolism , Stem Cell Factor/pharmacology , Humans , Macrophages/metabolism , Mice, Inbred C57BL
6.
Front Immunol ; 15: 1403789, 2024.
Article in English | MEDLINE | ID: mdl-39156897

ABSTRACT

Streptococcus suis causes diseases in pigs and has emerged as a zoonotic agent. When infected, the host develops an exacerbated inflammation that can lead to septic shock and meningitis. Although neutrophils greatly infiltrate the lesions, their dynamics during S. suis infection remain poorly described. Moreover, very few studies reported on the production and role of a key factor in the regulation of neutrophils: the colony-stimulating granulocyte factor (G-CSF). In this study, we characterized the G-CSF-neutrophil axis in the pathogenesis of S. suis induced disease. Using a mouse model of S. suis infection, we first evaluated the recruitment of neutrophils and their activation profile by flow cytometry. We found that infection provokes a massive neutrophil recruitment from the bone marrow to the blood and spleen. In both compartments, neutrophils displayed multiple activation markers. In parallel, we observed high systemic levels of G-CSF, with a peak of production coinciding with that of neutrophil recruitment. We then neutralized the effects of G-CSF and highlighted its role in the release of neutrophils from the bone marrow to the blood. However, it did not affect bacteremia nor the cytokine storm induced by S. suis. In conclusion, systemic G-CSF induces the release of neutrophils from the bone marrow to the blood, but its role in inflammation or bacterial clearance seems to be compensated by unknown factors. A better understanding of the role of neutrophils and inflammatory mediators could lead to better strategies for controlling the infection caused by S. suis.


Subject(s)
Granulocyte Colony-Stimulating Factor , Neutrophil Infiltration , Neutrophils , Streptococcal Infections , Streptococcus suis , Streptococcus suis/immunology , Animals , Granulocyte Colony-Stimulating Factor/metabolism , Streptococcal Infections/immunology , Mice , Neutrophils/immunology , Neutrophils/metabolism , Neutrophil Infiltration/immunology , Disease Models, Animal , Female , Mice, Inbred C57BL
7.
Commun Biol ; 7(1): 914, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075283

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) play key roles in intestinal inflammation. Olfactomedin 4 (OLFM4) is highly expressed in the colon and has a potential role in dextran sodium sulfate-induced colitis. However, the detailed mechanisms underlying the effects of OLFM4 on ILC3-mediated colitis remain unclear. In this study, we identify OLFM4 as a positive regulator of IL-22+ILC3. OLFM4 expression in colonic ILC3s increases substantially during intestinal inflammation in humans and mice. Compared to littermate controls, OLFM4-deficient (OLFM4-/-) mice are more susceptible to bacterial infection and display greater resistance to anti-CD40 induced innate colitis, together with impaired IL-22 production by ILC3, and ILC3s from OLFM4-/-mice are defective in pathogen resistance. Besides, mice with OLFM4 deficiency in the RORγt compartment exhibit the same trend as in OLFM4-/-mice, including colonic inflammation and IL-22 production. Mechanistically, the decrease in IL-22+ILC3 caused by OLFM4 deficiency involves the apoptosis signal-regulating kinase 1 (ASK1)- p38 MAPK signaling-dependent downregulation of RAR-related orphan receptor gamma (RORγt) protein. The OLFM4-metadherin (MTDH) complex upregulates p38/RORγt signaling, which is necessary for IL-22+ILC3 activation. The findings indicate that OLFM4 is a novel regulator of IL-22+ILC3 and essential for modulating intestinal inflammation and tissue homeostasis.


Subject(s)
Colitis , Interleukin-22 , Interleukins , Mice, Knockout , Animals , Mice , Interleukins/metabolism , Interleukins/genetics , Colitis/genetics , Colitis/chemically induced , Colitis/metabolism , Colitis/immunology , Colitis/pathology , Humans , Lymphocytes/immunology , Lymphocytes/metabolism , Mice, Inbred C57BL , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/genetics , Immunity, Innate , Inflammation/metabolism , Inflammation/genetics , Male , Glycoproteins
8.
Aging (Albany NY) ; 16(13): 10799-10812, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38967628

ABSTRACT

OBJECTIVE: This investigation seeks to elucidate the role of the Granulocyte Colony-Stimulating Factor (G-CSF) in the progression of hepatocellular carcinoma (HCC), as well as the impact of the substance on related signaling pathways within the disease matrix. METHODS: Nude mouse tumor-bearing assay was used to detect tumor progression. Levels of Mannose/CD68 and CD34/Mannose within these samples and the concentrations of Mannose and inducible Nitric Oxide Synthase (iNOS) in macrophages were quantified using immunofluorescence techniques. The angiogenic capability was assessed via tube formation assays, and protein expressions of G-CSF, Vascular Endothelial Growth Factor (VEGF), Transforming Growth Factor-beta (TGF-ß), Matrix Metalloproteinases 2 and 9 (MMP2/9), SH2-containing protein tyrosine phosphatase-2 (SHP-2), phosphorylated PI3K/total PI3K (P-PI3K/t-PI3K), phosphorylated AKT/total AKT (P-AKT/t-AKT), and phosphorylated mTOR/total mTOR (P-mTOR/t-mTOR) were measured through Western Blot analysis in both tumor tissues and macrophages. RESULTS: Administration of G-CSF resulted in a marked augmentation of tumor volume. Macrophage Mannose expression was significantly elevated upon G-CSF treatment, while iNOS levels were conspicuously diminished. G-CSF substantially enhanced the secretion of VEGF, TGF-ß, and MMPs in tumor tissues. Macrophage parameters, following incubation in G-CSF pre-treated conditioned medium, indicated enhanced tube-forming capabilities relative to the control, an effect mitigated by the introduction of specific inhibitors. Furthermore, the G-CSF group exhibited a notable reduction in SHP-2 expression, alongside a substantial elevation in the phosphorylation levels of the PI3K/AKT/mTOR pathway proteins across all tumor-bearing paradigms. CONCLUSION: G-CSF ostensibly facilitates the advancement of hepatocellular carcinoma by activating the PI3K/AKT/mTOR signaling cascade within Tumor-Associated Macrophages (TAM).


Subject(s)
Carcinoma, Hepatocellular , Granulocyte Colony-Stimulating Factor , Liver Neoplasms , Mice, Nude , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Animals , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , TOR Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Mice , Humans , Tumor-Associated Macrophages/metabolism , Neovascularization, Pathologic/metabolism , Cell Line, Tumor , Macrophages/metabolism , Macrophages/drug effects , Male
9.
Cell Rep ; 43(8): 114542, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39046877

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) is widely used to enhance myeloid recovery after chemotherapy and to mobilize hematopoietic stem cells (HSCs) for transplantation. Unfortunately, through the course of chemotherapy, cancer patients can acquire leukemogenic mutations that cause therapy-related myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML). This raises the question of whether therapeutic G-CSF might potentiate therapy-related MDS/AML by disproportionately stimulating mutant HSCs and other myeloid progenitors. A common mutation in therapy-related MDS/AML involves chromosome 7 deletions that inactivate many tumor suppressor genes, including KMT2C. Here, we show that Kmt2c deletions hypersensitize murine HSCs and myeloid progenitors to G-CSF, as evidenced by increased HSC mobilization and enhanced granulocyte production from granulocyte-monocyte progenitors (GMPs). Furthermore, Kmt2c attenuates the G-CSF response independently from its SET methyltransferase function. Altogether, the data raise concerns that monosomy 7 can hypersensitize progenitors to G-CSF, such that clinical use of G-CSF may amplify the risk of therapy-related MDS/AML.


Subject(s)
Granulocyte Colony-Stimulating Factor , Granulocytes , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells , Granulocyte Colony-Stimulating Factor/metabolism , Animals , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/drug effects , Mice , Granulocytes/metabolism , Granulocytes/drug effects , Mice, Inbred C57BL , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/drug therapy , Humans , Methyltransferases/metabolism , Methyltransferases/genetics
10.
Cell Rep ; 43(7): 114475, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38996072

ABSTRACT

Endomucin (EMCN) currently represents the only hematopoietic stem cell (HSC) marker expressed by both murine and human HSCs. Here, we report that EMCN+ long-term repopulating HSCs (LT-HSCs; CD150+CD48-LSK) have a higher long-term multi-lineage repopulating capacity compared to EMCN- LT-HSCs. Cell cycle analyses and transcriptional profiling demonstrated that EMCN+ LT-HSCs were more quiescent compared to EMCN- LT-HSCs. Emcn-/- and Emcn+/+ mice displayed comparable steady-state hematopoiesis, as well as frequencies, transcriptional programs, and long-term multi-lineage repopulating capacity of their LT-HSCs. Complementary functional analyses further revealed increased cell cycle entry upon treatment with 5-fluorouracil and reduced granulocyte colony-stimulating factor (GCSF) mobilization of Emcn-/- LT-HSCs, demonstrating that EMCN expression by LT-HSCs associates with quiescence in response to hematopoietic stress and is indispensable for effective LT-HSC mobilization. Transplantation of wild-type bone marrow cells into Emcn-/- or Emcn+/+ recipients demonstrated that EMCN is essential for endothelial cell-dependent maintenance/self-renewal of the LT-HSC pool and sustained blood cell production post-transplant.


Subject(s)
Cell Lineage , Hematopoiesis , Hematopoietic Stem Cells , Animals , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Mice , Mice, Inbred C57BL , Cell Movement , Fluorouracil/pharmacology , Humans , Granulocyte Colony-Stimulating Factor/metabolism , Cell Cycle , Endothelial Cells/metabolism
11.
Int J Mol Sci ; 25(11)2024 May 25.
Article in English | MEDLINE | ID: mdl-38891957

ABSTRACT

Lymphocyte collection by apheresis for CAR-T production usually does not include blood mobilized using granulocyte colony stimulating factor (G-CSF) due to the widespread knowledge that it causes a decrease in the number and functionality of lymphocytes. However, it is used for stem cell transplant, which is a common treatment for hematological malignancies. The growing demand for CAR therapies (CAR-T and NK-CAR), both in research and clinics, makes it necessary to evaluate whether mobilized PBSC products may be potential candidates for use in such therapies. This review collects recent works that experimentally verify the role and functionality of T and NK lymphocytes and the generation of CAR-T from apheresis after G-CSF mobilization. As discussed, T cells do not vary significantly in their phenotype, the ratio of CD4+ and CD8+ remains constant, and the different sub-populations remain stable. In addition, the expansion and proliferation rates are invariant regardless of mobilization with G-CSF as well as the secretion of proinflammatory cytokines and the cytotoxic ability. Therefore, cells mobilized before apheresis are postulated as a new alternative source of T cells for adoptive therapies that will serve to alleviate high demand, increase availability, and take advantage of the substantial number of existing cryopreserved products.


Subject(s)
Granulocyte Colony-Stimulating Factor , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Humans , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Colony-Stimulating Factor/metabolism , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Immunotherapy, Adoptive/methods , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/immunology , Hematopoietic Stem Cell Mobilization/methods , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals
12.
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
13.
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
14.
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
15.
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
16.
Cytotherapy ; 26(8): 797-805, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38625068

ABSTRACT

Neutrophils are the most frequent immune cell type in peripheral blood, performing an essential role against pathogens. People with neutrophil deficiencies are susceptible to deadly infections, highlighting the importance of generating these cells in host immunity. Neutrophils can be generated from hematopoietic progenitor cells (HPCs) and embryonic stem cells (ESCs) using a cocktail of cytokines. In addition, induced pluripotent stem cells (iPSCs) can be differentiated into various functional cell types, including neutrophils. iPSCs can be derived from differentiated cells, such as skin and blood cells, by reprogramming them to a pluripotent state. Neutrophil generation from iPSCs involves a multistep process that can be performed through feeder cell-dependent and feeder cell-independent manners. Various cytokines and growth factors, in particular, stem cell facto, IL-3, thrombopoietin and granulocyte colony-stimulating factor (G-CSF), are used in both methods, especially, G-CSF which induces the final differentiation of neutrophils in the granulocyte lineage. iPSC-derived neutrophils have been used as a valuable tool for studying rare genetic disorders affecting neutrophils. The iPSC-derived neutrophils can also be used for disease modeling, infection research and drug discovery. However, several challenges must be overcome before iPSC-derived neutrophils can be used therapeutically in transplantation medicine. This review provides an overview of the commonly employed protocols for generating neutrophils from HPCs, ESCs and iPSCs and discusses the potential applications of the generated cells in research and medicine.


Subject(s)
Cell Differentiation , Hematopoietic Stem Cells , Induced Pluripotent Stem Cells , Neutrophils , Humans , Induced Pluripotent Stem Cells/cytology , Neutrophils/metabolism , Neutrophils/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Colony-Stimulating Factor/metabolism
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL