Figure 4 - uploaded by Haroon I Sheikh
Content may be subject to copyright.
G-CSF inhibits IL-12/23 p40 production in DCs. DCs were pretreated for 15 min with GR-1 CM in a 1:1 ratio with fresh media and then stimulated with LPS (100 ng/mL) and IFN-(1000 U/mL), and IL-12 release into the media was measured 24 h later by ELISA. GR-1 CM inhibits LPS-induced IL-12 release (A). DCs were stimulated for 18 h with CM-the final 6 h in the presence of brefeldin A. Intracellular IL-12/23 p40 production was measured by flow cytometry. G-CSFR/ DCs produce significantly higher levels of IL-12/23 p40 after GR-1 CM stimulation than WT DCs (B). Antibody-mediated neutralization of G-CSF within the GR-1 CM, by incubation of anti-G-CSF (1 g/mL) with GR-1 CM for 1 h prior to the addition of CM to the DC culture, also enhanced IL-12/23 p40 stimulation (C). DCs were pretreated for 45 min with rG-CSF (50 ng/mL) and then treated with LPS (100 ng/mL). IL-12 mRNA was measured by qPCR. rG-CSF inhibits il12b (p40 subunit of IL-12) mRNA transcription, as measured by real-time qPCR, 6 h after treatment (D). Data are shown relative to the positive control (LPS alone) and normalized to gapdh. No il12b expression was detected in nontreated cells. IL-12 protein (p70) was measured by ELISA in the culture supernatant of LPS (100 ng/mL) IFN-(1000 U/mL)-treated DCs after 18 h of stimulation. rG-CSF inhibits IL-12 secretion (E). *P 0.05; **P 0.01; unpaired t test (B) or paired t test (C-E), where samples from the same experiment are paired. Representative data of least two (A) or three (B-E) independent experiments. Data shown as mean sem. 

G-CSF inhibits IL-12/23 p40 production in DCs. DCs were pretreated for 15 min with GR-1 CM in a 1:1 ratio with fresh media and then stimulated with LPS (100 ng/mL) and IFN-(1000 U/mL), and IL-12 release into the media was measured 24 h later by ELISA. GR-1 CM inhibits LPS-induced IL-12 release (A). DCs were stimulated for 18 h with CM-the final 6 h in the presence of brefeldin A. Intracellular IL-12/23 p40 production was measured by flow cytometry. G-CSFR/ DCs produce significantly higher levels of IL-12/23 p40 after GR-1 CM stimulation than WT DCs (B). Antibody-mediated neutralization of G-CSF within the GR-1 CM, by incubation of anti-G-CSF (1 g/mL) with GR-1 CM for 1 h prior to the addition of CM to the DC culture, also enhanced IL-12/23 p40 stimulation (C). DCs were pretreated for 45 min with rG-CSF (50 ng/mL) and then treated with LPS (100 ng/mL). IL-12 mRNA was measured by qPCR. rG-CSF inhibits il12b (p40 subunit of IL-12) mRNA transcription, as measured by real-time qPCR, 6 h after treatment (D). Data are shown relative to the positive control (LPS alone) and normalized to gapdh. No il12b expression was detected in nontreated cells. IL-12 protein (p70) was measured by ELISA in the culture supernatant of LPS (100 ng/mL) IFN-(1000 U/mL)-treated DCs after 18 h of stimulation. rG-CSF inhibits IL-12 secretion (E). *P 0.05; **P 0.01; unpaired t test (B) or paired t test (C-E), where samples from the same experiment are paired. Representative data of least two (A) or three (B-E) independent experiments. Data shown as mean sem. 

Source publication
Article
Full-text available
MΦs are important sensory cells of the innate immune system and regulate immune responses through releasing different combinations of cytokines. In this study, we examined whether cytokines released by MΦs in response to the probiotic bacterial strain GR-1 modulate the responses of DCs. The cytokine profile released by GR-1-treated MΦs was characte...

Similar publications

Article
Full-text available
In a double-blind, placebo-controlled, randomized study, 10 healthy men received either a single dose of 480 microg granulocyte colony-stimulating factor (G-CSF) or saline. Blood taken from the volunteers was stimulated with 10 microg/mL endotoxin and released cytokines were measured by enzyme-linked immunosorbent assay. Expression of G-CSF recepto...
Article
Full-text available
The mechanisms mediating hematopoietic stem and progenitor cell (HSPC) mobilization by G-CSF are complex. We have found previously that G-CSF-enforced mobilization is controlled by peripheral sympathetic nerves via norepinephrine (NE) signaling. In the present study, we show that G-CSF likely alters sympathetic tone directly and that methods to inc...
Article
Full-text available
The hypothesis that marrow-derived cells, and specifically proinflammatory proteins in those cells, play a critical role in the development of diabetes-induced retinopathy and tactile allodynia was investigated. Abnormalities characteristic of the early stages of retinopathy and allodynia were measured in chimeric mice lacking inducible nitric oxid...
Article
Full-text available
Hematopoietic development is orchestrated by gene regulatory networks that progressively induce lineage-specific transcriptional programs. To guarantee the appropriate level of complexity, flexibility, and robustness, these networks rely on transcriptional and post-transcriptional circuits involving both transcription factors (TFs) and microRNAs (m...
Article
Full-text available
Background: Filgrastim or methionyl-granulocyte colony-stimulating factor (Met-G-CSF), is a recombinant therapeutic protein widely used to treat severe neutropenia caused by myelosuppressive drugs in patients with nonmyeloid malignancies. In addition to its role in the regulation of granulopoiesis, treatment with G-CSF is considered the standard a...

Citations

... In the innate immune system, the most well-known function of G-CSF is in maturation and trafficking of neutrophils from the blood to the periphery [33]. One study suggested that macrophages also produce very high levels of G-CSF in response to bacteria that may also modulate dendritic cell responses, thus suggesting a role in the interaction between innate and adaptive immune cells [34]. Binding of G-CSF to its receptor leads to phosphorylation of JAK kinases and subsequent recruitment of STAT5 transcription and has been reported to be involved in the maturation of granulocyte/macrophage precursors of activated G-/GM-CSFR [35]. ...
Article
Granulocyte colony-stimulating factor (G-CSF) is a cytokine most well-known for maturation and mobilization of bone marrow neutrophils. Although it is used therapeutically to treat chemotherapy induced neutropenia, it is also highly expressed in some tumors. Case reports suggest that tumors expressing high levels of G-CSF are aggressive, more difficult to treat, and present with poor prognosis and high mortality rates. Research on this topic suggests that G-CSF has tumor-promoting effects on both tumor cells and the tumor microenvironment. G-CSF has a direct effect on tumor cells to promote tumor stem cell longevity and overall tumor cell proliferation and migration. Additionally, it may promote pro-tumorigenic immune cell phenotypes such as M2 macrophages, myeloid-derived suppressor cells, and regulatory T cells. Overall, the literature suggests a plethora of pro-tumorigenic activity that should be balanced with the therapeutic use. In this review, we present an overview of the multiple complex roles of G-CSF and G-CSFR in tumors and their microenvironment and discuss how clinical advances and strategies may open new therapeutic avenues.
... Reduced G-CSF production however, was observed in cells isolated from IBD patient tissue [101]. G-CSF-mediated mechanisms of action are being elucidated; using G-CSFR-deficient DCs, it was observed that GR-1-conditioned media induced significant IL-12/23 p40 production, which indicated that the G-CSF within the GR-1 conditioned media inhibited IL-12/23 p40 production [102]. This suggests that GR-1 can regulate immune responses through releasing G-CSF, and that G-CSF inhibits pro-inflammatory cytokine production via crosstalk between macrophages and dendritic cells. ...
Article
Full-text available
There is a growing body of evidence documenting probiotic bacteria to have a beneficial effect to the host through their ability to modulate the mucosal immune system. Many probiotic bacteria can be considered to act as either immune activators or immune suppressors, which have appreciable influence on homeostasis, inflammatory- and suppressive-immunopathology. What is becoming apparent is the ability of these probiotics to modulate innate immune responses via direct or indirect effects on the signaling pathways that drive these activatory or suppressive/tolerogenic mechanisms. This review will focus on the immunomodulatory role of probiotics on signaling pathways in innate immune cells: from positive to negative regulation associated with innate immune cells driving gut mucosal functionality. Research investigations have shown probiotics to modulate innate functionality in many ways including, receptor antagonism, receptor expression, binding to and expression of adaptor proteins, expression of negative regulatory signal molecules, induction of micro-RNAs, endotoxin tolerisation and finally, the secretion of immunomodulatory proteins, lipids and metabolites. The detailed understanding of the immunomodulatory signaling effects of probiotic strains will facilitate strain-specific selective manipulation of innate cell signal mechanisms in the modulation of mucosal adjuvanticity, immune deviation and tolerisation in both healthy subjects and patients with inflammatory and suppressive pathology.
... Augmented inflammation, tissuespecific (increased pro-IL-1β and TNF-α expression in liver) as well as systemic (increased plasma IL-β, IL-6, IL-17 and MCP-1 levels) were responsible for the observed mortality in this model (Figures 6 and 7). Further, secretion of an antiinflammatory cytokine, such as G-CSF that gets elevated after LPS challenge as a compensatory mechanism, 43,44 was impaired in APAP-LPS model of toxicity developed in this study. We propose that increased plasma G-CSF is a natural anti-inflammatory mechanism to counter-balance excessive inflammation by LPS. ...
Article
Full-text available
Acetaminophen (N-acetyl-para-aminophenol (APAP)) toxicity causes acute liver failure by inducing centrilobular hepatic damage as a consequence of mitochondrial oxidative stress. Sterile inflammation, triggered by hepatic damage, facilitates gut bacterial translocation leading to systemic inflammation; TLR4-mediated activation by LPS has been shown to have a critical role in APAP-mediated hepatotoxicity. In this study, we demonstrate significant protection mediated by chitohexaose (Chtx) in mice challenged with a lethal dose of APAP (400 mg/kg b.w.). Decreased mortality by Chtx was associated with reduced hepatic damage, increased peritoneal migration of neutrophils, decreased mRNA expression of IL-1β as well as inhibition of inflammasome activation in liver. Further, an alternate mouse model of co-administration of a sublethal doses of APAP (200 mg/kg b.w.) and LPS (5 mg/kg b.w.) operating synergistically and mediating complete mortality was developed. Overwhelming inflammation, characterized by increased inflammatory cytokines (TNF-α, IL-1β and so on) in liver as well as in circulation and mortality was demonstrable in this model. Also, Chtx administration mediated significant reversal of mortality in APAP+LPS co-administered mice, which was associated with reduced IL-1β in liver and plasma cytokines in this model. In conclusion, Chtx being a small molecular weight linear carbohydrate offers promise for clinical management of liver failure associated with APAP overdose.
... 184 This same group went on to a more detailed characterization of the effect of L. rhamnosus GR-1 on murine macrophages and found similarly that the presence of these cells resulted in lower levels of TNF-α, as well as GM-CSF, IL-6, and IL-12 and greatly increased G-CSF production. 185 ...
Chapter
From birth to death the human host immune system interacts with bacterial cells. Biofilms are communities of microbes embedded in matrices composed of extracellular polymeric substance (EPS), and have been implicated in both the healthy microbiome and disease states. The immune system recognizes many different bacterial patterns, molecules, and antigens, but these components can be camouflaged in the biofilm mode of growth. Instead, immune cells come into contact with components of the EPS matrix, a diverse, hydrated mixture of extracellular DNA (bacterial and host), proteins, polysaccharides, and lipids. As bacterial cells transition from planktonic to biofilm-associated they produce small molecules, which can increase inflammation, induce cell death, and even cause necrosis. To survive, invading bacteria must overcome the epithelial barrier, host microbiome, complement, and a variety of leukocytes. If bacteria can evade these initial cell populations they have an increased chance at surviving and causing ongoing disease in the host. Planktonic cells are readily cleared, but biofilms reduce the effectiveness of both polymorphonuclear neutrophils and macrophages. In addition, in the presence of these cells, biofilm formation is actively enhanced, and components of host immune cells are assimilated into the EPS matrix. While pathogenic biofilms contribute to states of chronic inflammation, probiotic Lactobacillus biofilms cause a negligible immune response and, in states of inflammation, exhibit robust antiinflammatory properties. These probiotic biofilms colonize and protect the gut and vagina, and have been implicated in improved healing of damaged skin. Overall, biofilms stimulate a unique immune response that we are only beginning to understand.
... G-CSF is primarily known for its role in the generation, mobilization and function of neutrophils [1][2][3] which are key innate immune cells protecting against invading microbes [4][5][6][7]. G-CSF also plays an important role in regulating both innate and adaptive immune responses through modulating activation of macrophages and dendritic cells (DCs) [8][9][10], and promoting generation of tolerogenic DCs [11] and regulatory T cells [12]. These dual modalities of G-CSF in immune regulation could be important in maintaining intestinal immune homeostasis, where both avid immune surveillance and tolerance against infiltrating commensal microbes are required. ...
... In the human bladder carcinoma cell line T24, GR-1 alone has little stimulatory effects in producing inflammatory cytokines and chemokines; however, potentiated E. coli-induced activation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and production of inflammatory cytokines [24,25]. GR-1 can also render anti-inflammatory effects on macrophages [26,27] and dendritic cells [28], and promotes the generation of regulatory T cells in humans [29]. To date, detailed immune responses and signaling mechanisms elicited by GR-1 remain largely unknown. ...
... We previously showed that GR-1 potently induces G-CSF but poorly induces TNF-α production in macrophages [28]. To further examine production of other cytokines induced by GR-1 and their levels in comparison to other stimuli, immortalized bone marrow-derived macrophages (BMDMs) were treated with live GR-1, LPS, PAM 2 CSK 4 and PAM 3 CSK 4 for 5 h, and the expression of 84 cytokines and chemokines were examined using the RT 2 Profiler PCR Array System Kit. ...
... Here, we showed that among 84 cytokines/chemokines examined, GR-1 uniquely and potently induced G-CSF ( Fig. 1 and Additional file 1: Table S1), and the production of G-CSF by GR-1 was as efficient as other known pro-inflammatory stimuli in both kinetics and amounts (Fig. 2). This observation is consistent with our previous studies in macrophages [26,28] and placental trophoblast cells that showed high levels of G-CSF production by GR-1 [22,23]. GR-1 at higher than 20 CFU/cell or after 12 h post-exposure was able to induce TNF-α, albeit at significantly lower levels than those induced by E. coli GR-12 or S. aureus (Fig. 2). ...
Article
Full-text available
Different species and strains of probiotic bacteria confer distinct immunological responses on immune cells. Lactobacillus rhamnosus GR-1 (GR-1) is a probiotic bacterial strain found in both the intestinal and urogenital tracts, and has immunomodulatory effects on several cell types including macrophages. However, detailed immunological responses and the signaling mechanism involved in the response are largely unknown. We examined the production of GR-1-induced cytokines/chemokines and signaling events in macrophages. Among 84 cytokines and chemokines examined, GR-1 discretely induced granulocyte colony-stimulating factor (G-CSF) mRNA at highest levels (>60-fold) without inducing other cytokines such as IL-1α, IL-1β, IL-6 and TNF-α (<5-fold). The toll-like receptor (TLR) 2/6-agonist PAM 2 CSK 4 , TLR2/1-agonist PAM 3 CSK 4 and TLR4-agonist lipopolysaccharide induced all of these inflammatory cytokines at high levels (>50-fold). The TLR2 ligand lipoteichoic acid activated all mitogen-activated kinases, Akt and NF-κB; whereas, GR-1 selectively activated extracellular regulated kinases and p38, NF-κB and Akt, but not c-Jun N-terminal kinases (JNKs) in a TLR2-dependent manner. Using specific inhibitors, we demonstrated that lack of JNKs activation by GR-1 caused inefficient production of pro-inflammatory cytokines but not G-CSF production. A secreted heat-labile protein-like molecule, 30–100 kDa in size, induced the preferential production of G-CSF. This study elucidated unique signaling events triggered by GR-1, resulting in selective production of the immunomodulatory cytokine G-CSF in macrophages.
... The strain, in high doses, increased the pro-Th1/Th17 cytokine levels (TNF-α, IL-1B, IL-12p70, IL-12p40, and IL-23), but only caused a low increase in IL-10 concentration, indicating a semi-maturation of the cells, and a tendency to a pro-inflammatory effect. On the contrary, other authors [207,208] have suggested that L. rhamnosus affected macrophages-dendritic cells interaction through a granulocyte colonystimulating factor (G-CSF) modulation directed to the IL-12/23 p40 response of the dendritic cells. It is important to point out also that vaginal microbiota affects the fetus [209][210][211]. ...
Article
Probiotics, defined as live microorganisms that, when administered in adequate amounts, confer a health benefit on the host, are considered a valid and novel alternative for the prevention and treatment of female urogenital tract infections. Lactobacilli, the predominant microorganisms of the healthy human vaginal microbiome, can be included as active pharmaceutical ingredients in probiotics products. Several requirements must be considered or criteria fulfilled during the development of a probiotic product or formula for the female urogenital tract. This review deals with the main selection criteria for urogenital probiotic microorganisms: host specificity, potential beneficial properties, functional specifications, technological characteristics and clinical trials used to test their effect on certain physiological and pathological conditions. Further studies are required to complement the current knowledge and support the clinical applications of probiotics in the urogenital tract. This therapy will allow the restoration of the ecological equilibrium of the urogenital tract microbiome as well as the recovery of the sexual and reproductive health of women. Copyright © 2015. Published by Elsevier B.V.
... In contrast, a high constitutive level of G-CSF is detected in the colon [29], and administration of G-CSF confers protective effects on colitis, pulmonary inflammation [30], neuronal inflammation, cell death [31, 32] , and cardiac ischemia/reperfusion injury [33]. Furthermore, G-CSF reduces TNF-and IL-12 production in myeloid cells [29, 34], and promotes the generation of tolerogenic DCs and T lympho- cytes [23,353637. These seemingly opposing effects suggest that G-CSF has tissue-and disease-specific roles. ...
Article
Full-text available
The G-CSF is best known for its activity in the generation and activation of neutrophils. In addition, studies on G-CSF(-/-) or G-CSFR(-/-) mice and BMC cultures suggested a role of G-CSF in macrophage generation. However, our understanding on the role of G-CSF in macrophage development is limited. Here, using in vitro BMC models, we demonstrated that G-CSF promoted the generation of Gr-1(high)/F4/80(+) macrophage-like cells in M-BMCs, likely through suppressing cell death and enhancing generation of Gr-1(high)/F4/80(+) macrophage-like cells. These Gr-1(high) macrophage-like cells produced "M2-like" cytokines and surface markers in response to LPS and IL-4/IL-13, respectively. Adoptive transfer of EGFP-expressing (EGFP(+)) M-BMCs showed a dominant, gut-homing phenotype. The small intestinal lamina propria of G-CSFR(-/-) mice also harbored significantly reduced numbers of Gr-1(high)/F4/80(+) macrophages compared with those of WT mice, but levels of Gr-1(+)/F4/80(-) neutrophil-like cells were similar between these mice. Collectively, these results suggest a novel function of G-CSF in the generation of gut-homing, M2-like macrophages.
... There is growing a body of evidence that administration of G-CSF exerts neuroprotective abilities in stroke (10,12). Additionally, G-CSF itself possesses the protective effects through other mechanisms, including inhibition of apoptosis, and anti-inflammatory effects (10,16). ...
... There is growing a body of evidence that administration of G-CSF exerts neuroprotective abilities in stroke (10,12). Additionally, G-CSF itself possesses the protective effects through other mechanisms, including inhibition of apoptosis, and anti-inflammatory effects (10,16). ...
Article
Based on the neuroprotective effect of either G-CSF or statins in various neurological disease models, the purpose of this study was to evaluate the superiority of combined therapy G-CSF with simvastatin in experimental intracerebral hemorrhage (ICH). MATERIAL and Primary ICH was induced in male Sprague Dawley rats. G-CSF (50μg/kg), simvastatin (2 mg/kg), combined G-CSF and simvastatin, or phosphate buffered saline was given at 24 hours post-ICH. Neurobehavioral outcomes were assessed in all rats. The pathological changes of neuronal ultrastructure were examined with transmission electron microscopy at the given time. Simultaneously, immunohistochemical labeling and TUNEL assay were performed. Co-administration of G-CSF with simvastatin significantly promoted functional recovery and expedited the recovery time. Transmission electron microscopy revealed that combination treatment significantly improved ultrastructural outcomes. Histological examination showed that the expressions of Brdu co-labeled with NSE and GFAP, Factor VIII were higher in combined treatment than in control group. Additionally, the number of cell apoptosis was higher in control group than in experimental groups and lowest in combination group. Our results indicated that combination treatment of stroke with G-CSF and simvastatin augments the neuroprotective effect in rats after ICH.
Article
The objective of this study was to determine the effect of probiotic Lactobacillus rhamnosus GR-1 supernatant (GR-1 SN) on lipopolysaccharide (LPS) induced preterm birth (PTB) and outputs of cytokines, chemokines and progesterone in pregnant CD-1 mice. PTB rates following intrauterine injection of LPS with and without prior GR-1 SN treatment were compared. Cytokines and chemokines in the maternal plasma, myometrium, placenta and amniotic fluid were examined with multiplex assay, and circulating maternal progesterone was measured with enzyme-linked immunoassay. Statistical significance was assessed using One-Way Analysis of variance (ANOVA) or ANOVA on ranks. Fetal sex ratios in mice that delivered preterm were compared with those that delivered at term following LPS and GR-1 SN treatments. GR-1 SN reduced LPS-induced PTB by 43%. GR-1 SN significantly decreased the LPS-induced production of IL-1β, IL-6, IL-12p40, TNFα, CCL4 and CCL5 in maternal plasma; IL-6, IL-12p70, TNFα, IL-17 and IL-13 in myometrium; IL-6, IL-12p70, and IL-17 in placenta and IL-6, TNFα, CCL3 and CCL4 in amniotic fluid. Maternal plasma progesterone was significantly reduced following LPS injection with and without GR-1 SN pretreatment. There was no difference in fetal sex ratios between mice that delivered preterm and those that did not after LPS and GR-1 SN treatments. The supernatant of probiotic Lactobacillus rhamnosus GR-1 attenuated LPS-induced inflammation and PTB in vivo. GR-1 SN may confer therapeutic benefits in preventing infection-associated PTB by controlling systemic and intrauterine inflammation.