ROCK1 Functions as a Suppressor of Inflammatory Cell Migration by Regulating PTEN Phosphorylation and Stability.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 240-240
Author(s):  
Sasidhar Vemula ◽  
Jianjian Shi ◽  
Philip Hanneman ◽  
Lei Wei ◽  
Reuben Kapur

Abstract Abstract 240 Neutrophils and macrophages are major cellular components of the innate immune response and are recruited rapidly in large numbers to sites of infection. The small family of Rho GTPases and its downstream effectors, Rho kinases (Rho-associated, coiled-coil containing protein kinase) have been implicated in regulating various cellular functions including actin cytoskeleton organization, cell adhesion, and cell motility in non-hematopoietic cells. Rho kinases (ROCK1 and ROCK2) belong to a family of serine/threonine kinases whose role in inflammation is not known. Here we show that deficiency of ROCK1 but not ROCK2 results in increased recruitment of macrophages (3.2 fold, n=8, *p<0.01) and neutrophils (3.4 fold, n=5 *p<0.05) compared to WT controls in an in vivo model of aseptic peritonitis. In vitro, deficiency of ROCK1 in bone marrow derived macrophages shows a significant increase in haptotactic transwell migration in response to M-CSF as well as MCP-1 on fibronectin as well as an increase in migration towards the wounded area in a wound healing assay compared to controls (∼3 fold, n=3, *p<0.005). Consistently, deficiency of ROCK1 in bone marrow derived neutrophils also shows a ∼2.63 fold increase in migration in response to fMLP compared to WT bone marrow derived neutrophils (BMNs) in a chemotactic migration assay. ROCK1 deficient macrophages also demonstrate a ∼2.5 fold increase in adhesion on fibronectin (n=3, *p<0.002). The enhanced migration and adhesion in ROCK1−/− macrophages was observed in spite of comparable expression of F4/80 (WT; 85.63% vs. ROCK1−/−; 88.68%, n=4), α4β1 and α5β1 integrins (WT; 67.49% & 88.2% vs. ROCK1−/−; 71.82 % & 87.09%, n=4), while no significant difference in the phagocytosis of sheep red blood cells was observed between WT and ROCK1−/− macrophages (Phagocytic index: WT; 98% vs. ROCK1−/− 97%, n=3, p>.05). Close examination of the cytoskeleton of ROCK1 deficient macrophages using confocal microscopy revealed more F-actin content on the entire cell surface compared to wildtype controls. Consistently, flow cytometric analysis using Alexa 488-phalloidin staining revealed abundance of F-actin in ROCK1−/− macrophages compared to WT controls (WT; 46.19% vs. ROCK1−/−; 65.23%, n=3, *p<0.05). Furthermore, immunofluorescence imaging of podosomes carried out using anti-vinculin antibody revealed more pronounced and increased podosomes in ROCK1 deficient macrophages compared to WT controls (n=3, *p<0.05). Biochemical analysis of ROCK1−/− macrophages revealed that the enhanced recruitment of ROCK1 deficient macrophages and neutrophils was apparent in spite of normal expression of ROCK2 in ROCK1−/− cells and a 60% reduction in overall ROCK activity. Interestingly, although both ROCK1 and ROCK2 co-immunoprecipitate with PTEN in response to cytokine induced stimulation, only ROCK1 appeared to be essential for PTEN phosphorylation, activation and stability. In the absence of ROCK1, PTEN phosphorylation, its activity and stability were significantly impaired in spite of the presence of ROCK2 (n=3, *p<0.05). Consequently, an increase in the activation of downstream targets of PTEN including AKT, GSK-3β and cyclinD1 was observed in ROCK1 deficient macrophages relative to controls (n=3). Taken together, these studies reveal a biochemical pathway involving ROCK1 and PTEN which is involved in the recruitment of macrophages and neutrophils during acute inflammation. Thus, ROCK1 likely functions as a physiologic regulator of PTEN whose function is to repress excessive recruitment of macrophages and neutrophils during acute inflammation. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2406-2406
Author(s):  
Sasidhar Vemula ◽  
Premchand Gandra ◽  
Jianjian Shi ◽  
John T. O’Malley ◽  
Ayek-Nati N. Ahyi ◽  
...  

Abstract Neutrophils and macrophages are a major cellular component of innate immune response and are recruited rapidly in large numbers to sites of infection. The small GTPase Rho and its downstream effectors, the serine/threonine kinase ROCKI and ROCKII have been implicated in various cellular functions including actin cytoskeleton organization, cell adhesion, and cell motility in non-hematopoietic cells. We show that loss of ROCKI results in a significant reduction in the number of B lymphocytes in the bone marrow (BM). The number of late pre-B cells were significantly reduced in ROCKI−/− bone marrow compared to WT controls (n=4, WT [51%] vs. ROCKI−/− [40%]). No significant difference in the number of pro/pre B-cells or immature B cells were observed in ROCKI−/− BM compared to WT controls (n=4). Surprisingly, recirculating mature B cells were significantly increased in ROCKI−/− BM compared to WT (n=4, WT [13.25%] vs. ROCKI−/− [25.2%]). Consistently, in vitro migration of ROCKI−/− B lymphocytes towards SDF-1was also significantly reduced compared to WT controls. Next, we analyzed the recruitment of macrophages deficient in the expression of ROCKI using a well-studied model of aseptic thioglycolate-induced peritonitis. In spite of comparable expression of F4/80 (WT; 85.63% vs. ROCKI−/−; 88.68%, n=4) and α4β1 and α5β1 integrins (WT; 67.49% & 88.2% vs. ROCKI−/−; 71.82% & 87.09%, respectively, n=4) in WT and ROCKI−/− macrophages, a ∼3.2 fold increase in the migration of macrophages was observed in the peritoneal cavity of ROCKI−/− mice compared to controls (WT; 0.46 X 106 vs. ROCKI−/−; 1.5 × 106, n=3, p&lt;0.01). In vitro, ROCKI deficiency in macrophages resulted in significant increase (∼2.2 fold) in haptotactic transwell migration in response to M-CSF on fibronectin coated wells (n=3, *p&lt;0.0075) as well as an increase in migration in a wound healing assay compared to controls (∼3 fold, n=3, *p&lt;0.005). In addition, ROCKI deficient macrophages also demonstrated a ∼2.5 fold increase in adhesion and spreading on fibronectin (n=3, *p&lt;0.002), although no significant difference in the phagocytosis of sheep red blood cells was observed between WT and ROCKI−/− macrophages (Phagocytic index: WT; 98% vs. ROCKI−/− 97%, n=3, p&gt;.05). Close examination of the cytoskeleton of ROCKI deficient macrophages using confocal microscopy revealed that F-actin was localized more across the entire cell surface in spread cells compared to wildtype controls. Consistently, flow cytometric analysis using Alexa 488-phalloidin staining revealed abundance of F-actin in ROCKI−/− macrophages compared to WT controls (WT; 46.19% vs. ROCKI−/−; 65.23%, n=3, *p&lt;0.05). Furthermore, immunofluorescence imaging of focal adhesion complexes carried out using anti-vinculin antibody revealed more pronounced and increased focal adhesion sites in ROCKI deficient macrophages compared to WT controls. Biochemically, deficiency of ROCKI resulted in increase levels of cyclinD1, a molecule known to play a critical role in macrophage migration. Consistently, deficiency of ROCKI in neutrophils also demonstrated a ∼2.63 fold increase in migration in response to fMLP in vitro and in vivo in response to thioglycolate challenge compared to WT controls (WT; 0.47 X 106, ROCKI−/− vs. 1.6 × 106, *p&lt;0.05). Taken together, our results provide a critical physiologic role for ROCKI in regulating both macrophage and neutrophil functions.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2642-2642
Author(s):  
Stephen B Ting ◽  
Sara Rhost ◽  
Sarah Ghotb

Abstract Despite the relative rarity of haematopoietic stem cells (HSCs) within the blood system, functional heterogeneity is paramount to their ability to sustain lifelong blood production. The quiescent HSC sits at the functional apex possessed with self-renewal properties and the greatest repopulation output. We previously identified the gene, Ap2a2 as an enhancer of HSC function and its protein as a potential cell fate determinant in HSC asymmetric cell divisions (Ting SB et al., Blood 2012). Mechanistically, we hypothesise Ap2a2 induces a state of HSC quiescence. Using the Tet-On histone H2B-GFP mouse model (Foudi et al., Nat Biotech 2009), we have shown Ap2a2 to be highly and differentially expressed in the predominantly, G0 dormant CD150+48-LSK GFPhigh as opposed to the more cycling GFPlow HSC subpopulation. Competitive transplantation of Ap2a2- versus empty vector-transduced H2B-GFP HSCs results in a three-fold increase of the CD150+48-LSK GFPhigh HSC subpopulation. To further confirm the importance of Ap2a2 in haematopoiesis, we have constructed Ap2a2-LacZ reporter and constitutive Ap2a2 knockout (KO) mouse lines. The Ap2a2 LacZ reporter with b-galactosidase flow cytometry staining of bone marrow subpopulations confirmed high endogenous Ap2a2 expression in the CD150+48-LSK long-term (LT-) versus CD150-48-LSK short-term (ST-) repopulating HSCs. Interim analyses of the constitutive Ap2a2 KO mice have revealed two obvious phenotypes: 14% of Ap2a2-null mice termed "non-survivors" are smaller, paler with failure of fetal liver (FL) development and die between E18.5 and weaning, whilst the remaining 11% are adult viable "survivors". However, at E14.5, Ap2a2-null compared to Ap2a2-wild type fetal livers showed less absolute total FL cells but increased CD150+48-LSM FL HSCs. This was quantitatively correlated via limiting dilution assay assessed at 16 weeks post-transplant with a two-fold increase in Ap2a2-null HSC numbers (1 in 78,917 versus 1 in 150,891, p=0.027). This suggests Ap2a2 has a role in FL HSC differentiation and/or fate with potential impairment of symmetrical versus asymmetrical HSC divisions currently being studied. When E14.5 FL cells were competitively transplanted, the Ap2a2-null HSC had impaired donor reconstitution function measured at 16 weeks post-transplant (19.8% versus 48.6%, p=0.015). Ap2a2-null versus wild-type E14.5 FL cells showed equivalent numbers of primary in vitro methylcellulose colony assays but loss of secondary colonies upon re-plating indicative of loss of in-vitro HSC self-renewal. Importantly, although the Ap2a2 adult "survivors" exhibited normal quantities of bone marrow HSC subpopulations, when functionally assessed, Ap2a2-null adult "survivor" HSCs showed loss of in-vivo HSC self-renewal in secondary transplantation assays. To investigate potential cellular mechanisms, we studied the cell cycle state of Ap2a2-null and wild-type E14.5 FL cells and identified that Ap2a2-null "non-survivors" had a relative loss of quiescent G0, specifically in the LT-HSC (and not seen in the ST-HSC) subpopulation throughout all of (E14.5 to E18.5) FL development. In contrast, the LT-HSC subpopulation in FLs of Ap2a2-null "survivors" had an initial loss of G0 at E14.5 but a compensatory increase in LT-HSC G0 by E18.5. Our preliminary data suggests Ap2a2 is a crucial factor for the quiescent LT-HSC subpopulation, and we propose that both during the highly proliferative fetal liver stage of haematopoiesis and adult HSCs under stress that Ap2a2 maintains a critical balance of dormant ("deep-sleeper") HSCs to ensure global HSC function. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3628-3628
Author(s):  
Marina Prewitz ◽  
Friedrich Philipp Seib ◽  
Martin Bornhaeuser ◽  
Carsten Werner

Abstract Abstract 3628 Poster Board III-564 The bone marrow (BM) harbours haematopoietic stem/progenitor cells (HSCs) in anatomically distinct sites (niches) where HSCs are subjected to regulatory cues such as cytokines, cell-cell contacts and extra-cellular matrix (ECM) all of which control stem cell fate. In particular mesenchymal stromal cells (MSCs) are an integral part of the bone marrow and are known to be key regulators of the HSC niche. We have previously shown that bio-artificial scaffolds can have a significant impact on the in vitro behaviour of MSCs. Here, we are therefore focussing on the role of (native) ECM within the MSC-HSC microenvironment by building on our previous findings and published data (Seib et al.,Tissue Eng Part A., 2009 in press). Thus the aim of the current study is (a) to identify niche-specific ECM components and (b) the use of such ECMs for in vitro culture of BM-derived stem cells. To mimic the natural ECM composition of the BM, different ECM types were generated from BM-derived cells using (a) Dexter cultures, (b) standard MSC cultures, (c) MSCs subjected to osteogenic differentiation. After 10 days of culture those MSC-derived ECMs were decellularised using 0.5% Triton-X and 20mM NH4OH leaving only the ECM behind (verified by scanning electron microscopy). Those ECMs were used as a substrate for a second culture of MSCs, which were analysed for their proliferation and differentiation potential. Cell-free ECM from standard MSC cultures improved MSC proliferation compared to cells grown on regular tissue culture plastic (TCP) over the period of 8 days. Most notably, all cell-free ECM preparations lead to a significant difference in the cytoskeletal arrangement of MSCs during the first 2 days of culture compared to TCP controls. Cultivation of MSCs on native ECM provided a guiding structure for those cells to grow into, and helped to maintain an elongated cell shape compared to substantial cell spreading on TCP (roundness 0.2 versus 0.5 and cell area of 2.2 versus 8.2mm2, respectively, p<0.001, n=60. A factor of 1 was set to equate to a perfect circle). Next, we investigate if native ECM could either directly improve HSC cultures or maximise MSC feeder characteristics. For the latter set of studies MSCs were initially cultured for 7 days on cell-free ECM (from standard MSC cultures) and subsequently co-cultured with human peripheral blood CD34+ HSCs in serum free medium supplemented with cytokines (Tpo, Flt3, and SCF at 10ng/ml). Following a 14 day culture period up to 3.5-fold more CD34+ cells were present in ECM co-cultures compared to TCP co-cultures that was accompanied with an overall expansion of CD45+ cells of 109-fold versus 35-fold, respectively. Our data suggest that ECM preparations derived from MSCs might be useful to accomplish better expansion of HSCs under defined culture conditions. In addition, this system permits the identification of bimolecular key components that can be utilized in the future design of simple and robust carrier systems for improved HSC maintenance in vitro. Figure HSC-MSC co-culture on preformed ECM substrates. (A) MSC-derived ECM (from standard MSC culture) following cell lysis (complete absence of cells). (B) Growth of a new set of MSCs on ECM substrates as shown in (A). (C) HSC-MSC co-culture on ECM substrates. Scale bars at 2μm. Arrow heads point out ECM structures. Figure HSC-MSC co-culture on preformed ECM substrates. (A) MSC-derived ECM (from standard MSC culture) following cell lysis (complete absence of cells). (B) Growth of a new set of MSCs on ECM substrates as shown in (A). (C) HSC-MSC co-culture on ECM substrates. Scale bars at 2μm. Arrow heads point out ECM structures. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2620-2620
Author(s):  
Christian Saure ◽  
Fabian Zohren ◽  
Thomas Schroeder ◽  
Ingmar Bruns ◽  
Ron Patrick Cadeddu ◽  
...  

Abstract Abstract 2620 Introduction: In our previous report (Zohren et al., Blood 2008) we could show that the blockade of the heterodimer VLA-4 by the monoclonal IgG4 antibody natalizumab leads to a significant increase in circulating CD34+ cells in patients with multiple sclerosis (MS). We now extend our analysis on the influence of natalizumab on CD34+ cells comparing bone marrow (BM) and peripheral blood (PB) derived CD34+ cells of natalizumab patients with those from healthy donors. Methods: A total of 83 patients with MS receiving natalizumab were included. In vitro adhesion, migration and apoptosis assays as well as LTC-IC of immunomagnetically enriched CD34+ cells were conducted. Flow cytometric analyses were performed to assess phenotype and composition of the CD34+ subsets. Results: The median concentration of circulating CD34+ cells was significantly greater compared to normal donors (7.7/μL vs. 1.8/μ L; p= 0.0001) and remained relatively stable during a one year treatment with natalizumab. Leukocyte cell counts, the number of T cell subsets as well as the number of CD19+ B cells and CD56+ natural killer cells were in normal range in PB and BM after short- and long-term treatment with natalizumab. However, we found significantly reduced adhesion and migration abilities of circulating CD34+ cells under natalizumab treatment in comparison to G-CSF mobilized CD34+ cells of healthy donors. Moreover, the self-renewal capacity of these cells was poor. In contrast, no significant difference was seen between the BM of natalizumab patients and the BM of healthy donors with regard to cellularity and proportion of CD34+ cells. In addition, neither co-expression of CD49d nor the adhesion ability of the BM derived CD34+ cells revealed a significant difference between the two collective. Conclusions: Our data indicate that natalizumab mediates an increase in circulating CD34+ cells by impaired homing. These findings argue against the use of natalizumab-exposed PB CD34+ cells for transplantation. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3859-3859
Author(s):  
Phuong L. Doan ◽  
J. Lauren Russell ◽  
Heather A. Himburg ◽  
Sarah K. Meadows ◽  
Pamela Daher ◽  
...  

Abstract Abstract 3859 Hematopoietic stem cell (HSC) regeneration is influenced by specialized bone marrow (BM) microenvironments, but the mechanisms that drive HSC regeneration remain incompletely defined. We have recently reported that deletion of the pro-apoptotic proteins, Bak and Bax, in Tie2+ bone marrow endothelial cells (BM ECs)(Tie2Cre;Bak-/-;BaxFl/- mice) caused a significant protection of the BM HSC pool and the BM sinusoidal vasculature in mice following high dose total body irradiation (TBI). We also confirmed that this protection of the BM HSC pool was caused by protection of BM Tie2+ ECs via generation of chimeric mice (Tie2Cre;Bak-/-;BaxFl/- BM; wild type BM ECs) which contained 4.8-fold less BM long-term repopulating HSCs compared to mice bearing deletion of Bak and Bax in both BM HSCs and BM ECs. In order to determine the mechanism through which Tie2+ BM ECs regulate HSC regeneration, we generated primary BM EC lines from Tie2Cre;Bak-/-;BaxFl/- mice and Tie2Cre;Bak-/-;BaxFl/+ control mice. We then compared the capacity for Bak/Bax -/- BM ECs to support BM HSC regeneration in vitro compared to Bak/Bax +/&minus; BM ECs. BM c-kit+sca-1+lin- (KSL) stem/progenitor cells were irradiated with 300 cGy and then placed in 7 day culture with Bak/Bax -/- BM ECs or Bak/Bax +/&minus; BM ECs. Culture with Bak/Bax -/- BM ECs did not yield a significant increase in total viable cells, but yielded 2000-fold increased number of BM KSL cells (p < 0.05, n=3) compared to cultures with Bak/Bax +/&minus; ECs. This significant expansion of phenotypic BM stem/progenitor cells corresponded to a 4-fold increase in CFU-S12 cells in the Bak/Bax -/- EC cultures vs. Bak/Bax +/&minus; EC cultures (p=0.01, n=5). We subsequently compared the level of expression of several microenvironmental ligands which are putatively involved in regulating hematopoiesis. We found that BM ECs from Tie2Cre;Bak-/-;BaxFl/- mice had 37-fold lower expression of stromal-derived factor-1 (SDF-1, CXCL12) compared to BM ECs from Tie2Cre;Bak-/-;BaxFl/+ mice. Moreover, 7 days after TBI, Tie2Cre;Bak-/-;BaxFl/- mice had a 41-fold increase in total viable BM cell counts and had a persistently lower SDF-1 expression on BM ECs (2.7-fold) compared to Tie2Cre;Bak-/-;BaxFl/+ mice (p=0.003). Therefore, we hypothesized that inhibition of SDF-1 signaling might facilitate hematopoietic regeneration following injury. Interestingly, the addition of a blocking anti-SDF1 antibody to cultures of irradiated BM KSL cells with Bak/Bax -/- ECs caused a 50% increase in total cell recovery (p<0.05), a 2.5 fold increase in BM KSL cell recovery (p<0.05) and a 2.2-fold increase in BM CFC recovery (p<0.05) compared to culture with Bak/Bax -/- ECs alone. However, the addition of anti-SDF1 antibody caused a 3-fold decrease in CFU-S12 recovery compared to Bak/Bax -/- EC cultures without anti- SDF1 antibody (p<0.05). Taken together, these data suggest that inhibition of SDF-1 signaling via BM ECs accelerates BM progenitor cell regeneration following injury but is deleterious to the recovery of the BM HSC pool. Targeted therapies aimed at inhibition of SDF-1 signaling may facilitate short-term hematopoietic reconstitution following injury via modulation of BM vascular niche signaling, but this may be at the expense of the BM HSC pool. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2837-2837 ◽  
Author(s):  
Ajay Kundra ◽  
Stacey Baptiste ◽  
Chi Chen ◽  
Hemant Sindhu ◽  
Jen-Chin Wang

Abstract PD-1 and MDSC are two recently discovered important tumor immune escape mechanisms. These immune escape pathways have been studied and their levels were found to be elevated in tumors such as myeloma, pancreatic, renal, ovarian, and breast cancers. Therefore, we studied these two immune escape pathways in patients with myeloid neoplasms including polycythemia vera (PV), Essential thrombocythemia (ET), myelofibrosis (MF) including primary myelofibrosis (PMF) and Post-ET, Post-PV myelofibrosis. We also studied IMID drug in vitro to see if IMID drug mechanism is related to these two escape pathways. 51 patients with MPN and 15 normal volunteer controls were studied. For quantification of expression of PD-1 and PD L-1 in MNC subpopulations (CD4+, CD8+ or CD14+ cells). Ficoll-paque- isolated MNCs were stained with PD-1(CD279-APC) and PD L-1 (CD274-PE)BD Biosciences; CA) plus either CD4-FITC, CD8-FITC or CD14-FITC, along with 7AAD (6 ug/ml), and subjected to flow cytometric analysis. The flow data was analyzed using FlowJo (v 7.6.2). PD-1 or PD L-1 positive cells were expressed as percentage in the subpopulation of cells assayed. For MDSC's, after Ficoll-Paque density centrifugation of peripheral blood, 106 mononuclear cells were stained for flow cytometric analysis using CD11b-APC, CD33-PE, and CD14-FITC (BD Biosceinces; Carlsbad, CA), along with their matched isoptoye controls. 7AAD (6 ug/ml) was used to exclude dead cells to eliminate nonspecific antibody binding. 50,000 events per specimen were acquired and the resultant flow cytometric data was analyzed by FlowJo software. MDSC's were defined as CD11b+CD14-CD33+ cells and were calculated as percentage of viable gated MNC's. MDSC's were also tested for in vitro inhibitory effects on Tcells : MDSC cell sorting was performed by flow cytometry of CD33+ cells from CD11b+CD14- microbead selected cells. CD3 cells were then labeled with CFSE (Life Technologies; CA) and incubated with and without the sorted MDSC's in the presence of CD3+CD28+ microbeads for five days before assessing CFSE activity. For IMID drug in vitro experiments, Pomalidomide (Pm)( Celgene) (10ϰg/ml) was added to mononuclear cells in the presence of CD3+CD28+ microbeads in culture medium. After 5-7 days, cells were assayed for PD-1, PD-L1 expression and MDSC's. Since our data showed no significant difference between ET, PV and MF, we grouped ET, PV and MF together as MPN. The results showed that there is no significant difference between patients with MPN and controls of PD-1 and PD-L1 expression in CD4+, CD8+ and CD14+ cells. However, MDSC's were significantly elevated in patients with MPN compared to controls (Fig 1.). Pomalidomide significantly reduced the expression of PD-1 and MDSC's- expressed as fold change of Pm/DMSO (mean+ SE) in CD4 +,( 0.46 ± 0.120, p= 0.05) , CD8+ (0.66±0.07,p=0.02), MDSC ( 0.71 ± 0.09, P=0.01). Pomalidomide has no significant effect on PD-1 expression in CD14+ cells and PD1-L-1 expression in CD4+, CD8+ and CD14 + cells .Sorted MDSC's significantly suppressed proliferation of the CFSE labeled CD3+ T cells. We conclude that 1) PD-1, PD L-1 expression is not altered in MPN patients. However, MPN patients have significantly increased MDSC's. 2) Pomalidomide, in vitro, significantly reduces the expression of PD-1 in CD4+, CD8+ cells and decreases MDSC. This data may be added to be one of the important mechanisms of immunomodulatory drugs in the treatment of Myelofibrosis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4717-4717
Author(s):  
Zonghong Shao ◽  
Xifeng Dong ◽  
Rong Fu

Objective To compare the response of GPI-AP negative or positive bone marrow mononuelear cells(BMMNCs) from PNH patients to DAG in vitro and explore the related mechanism. Methods Seventeen PNH patients as well as fourteen normal controls were enrolled. CD59-/CD59+ cells were sorted by magnetic activated cell-sorting system. Then the cells were incubated in IMDM medium containing several hemapoitic growth factors with DAG or G-CSF for 48h in vitro. The cell cycle kinetics and apoptosis of these cells were detected by flow cytometry(FCM). The expressions of CD114 on CD34+CD59- and CD34+CD59+ bone marrow cells(BMC) after incubated with G-CSF were measured by FCM. And another 14 PNH peripheral blood samples were obtained, the expression of CD44/CD49d on CD59- and CD59+ cells were analyzed by FCM respectively. The mRNA of CD114 and CD44/CD49d was also tested in 22 PNH patients vs 14 controls and CD59- vs CD59+ cells from 14 PNH patients by Q-PCR. Results After incubated with DAG for 48h in vitro, the death rate and apoptosis rate for GPI-AP negative and positive cells(CD59-/CD59+ BMMNCs cells): for CD59- BMMNCs, compared with control group, the death rate of DAG group increased (27.29±22.04% vs 19.10±20.93%), apoptosis rate also increased(10.55±12.34% vs 7.2±6.76%), there was no significant difference for them; for CD59+ BMMNCs, compared with control, the death rate of cells from DAG group increased significantly (31.89±26.75% vs 12.83±18.92%)(P<0.05), whereas there was no significant difference for the apoptosis rate (9.66±7.96% vs 6.31±1.32%); for the CD59- and CD59+ BMMNCs, the death rate was significant higher than apoptosis rate respectively(P<0.05). For cell cycle kinetics, there was no significant difference between the two kinds of BMMNCs. As to the percentage of CD114, compared with control group, it increased significantly in CD34+CD59+ BMMNCs from G-CSF group (48.12±41.20% vs 12.84±15.32%) (P<0.05), whereas there was no significant difference for CD34+CD59- BMMNCs (41.76±44.62% vs 26.79±41.62%). And the variation of CD114 for CD34+CD59+ BMMNCs was higher than that for CD34+CD59- BMMNCs(33.97±36.03% vs 14.88±27.02%)(P<0.05). The expression of CD44/CD49d protein: the expression of CD44 for CD59+was higher than that for CD59- cells(97.66±4.21% vs 93.46±9.52%, P<0.05); and there was no significant difference for CD49d expression in CD59- and CD59+ cells(38.46±27.37% vs 43.79±24.77%). The mRNA expressions of CD114, CD44 and CD49d in 22 PNH patients compared with 15 control and CD59- cells compared with CD59+ cells from 14 PNH patients : for CD114, its mRNA expression was higer for CD59+ cells compared with that for CD59- cells(2.78±2.52 vs 1.69±2.34, P<0.05), but there was no significant difference for CD114 in PNH patients and controls; for CD44, the significant difference exited for PNH patients compared with controls and CD59- cells compared with CD59+ cells(1.73±2.20 vs 3.80±3.87, P<0.05; 0.82±0.75 vs 2.38±2.42, P<0.05); for CD49d, no significant difference exited for PNH patients compared with controls and CD59- cells compared with CD59+ cells(2.83±2.62 vs 2.56±3.04; 1.74±2.60 vs 1.94±3.02). Conclusions In vitro, effect of DAG was similar on CD59- and CD59+ BMMNCs, the style of death was necrosis not apotosis and the cell cycle was not influenced by DAG. The variation of CD114 for CD34+CD59- after G-CSF stimulation was less than that in CD34+CD59+ cells, and mRNA of CD114 was lower in CD59- cells compared with CD59+ cells, which may indicating the mechanism for the remission of PNH patients after DAG chemotherapy. The protein and mRNA of CD44 was lower in PNH patients and CD59- cells compared with control and CD59+ cells respectively, which may explain the inferior growth of PNH cells, because they can not fully use the BM microenvironment. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4962-4962
Author(s):  
Hyejoo Park ◽  
Daeyoon Kim ◽  
Chansu Lee ◽  
Eunkyung Bae ◽  
Kwang-Sung Ahn ◽  
...  

Abstract Various factors released from BMSCs regulate the biologic behaviors of AML cells involving in responsiveness of chemotherapeutic treatment. Cytotoxic effect of Ara-C decreased when AML cells co-cultured with BMSCs. IL-6 expression was prominently increased when HEL cells were co-cultured with BMSCs. Even though IL-6 did not affect the growth of HEL cells, Ara-c mediated apoptosis was suppressed by BMSCs. Also, IL-6 induced the phosphorylation of AKT and its downstream gene (mTOR). Ara-C mediated H2AX mRNA was suppressed when Ara-C was treated with co-cultured HEL cells with BMSCs. Also, its expression was down-regulated in HEL cells co-treated with Ara-C plus 25 nM IL-6. Prevention of IL-6 mediated signaling by gp130 shDNA slightly suppressed Ara-C induced H2AX expression when gp130 shDNA transfected HEL cells were co-cultured with BMSCs under 10-6 M Ara-C treatment. In vivo model, we found that IL-6 expression levels in serum of mice detecting AML cells following Ara-C treatment were higher than in serum of mice not detecting residual AML cells. Even though somatic mutation of gp130 gene was not detected in the genome analysis of AML, the overall survival was statistically different depending on the IL-6 levels in serum of bone marrow. Our findings suggest that IL-6 releasing from BMSCs help AML cells to survive against Ara-C treatment resulting in developing relapase from enhancing the growth of minimal residual cells. Figure 1. IL-6 suppresses Ara-C mediated apoptosis of HEL in vitro assay. Figure 1. IL-6 suppresses Ara-C mediated apoptosis of HEL in vitro assay. Figure 2. IL-6 suppresses Ara-C mediated H2AX expression in HEL cells. Figure 2. IL-6 suppresses Ara-C mediated H2AX expression in HEL cells. Figure 3. Survival curve depending on IL-6 level of AML patients bone marrow serum. Figure 3. Survival curve depending on IL-6 level of AML patients bone marrow serum. Disclosures No relevant conflicts of interest to declare.


Author(s):  
Oliver J. Herd ◽  
Gulab Fatima Rani ◽  
James P Hewitson ◽  
Karen Hogg ◽  
Andrew P Stone ◽  
...  

Immune thrombocytopenia (ITP) is an acquired autoimmune condition characterized by both reduced platelet production and the destruction of functionally normal platelets by sustained attack from the immune system. However, the effect of prolonged ITP on the more immature hematopoietic progenitors remains an open area of investigation. Using a murine in vivo model of extended ITP, we reveal that ITP progression drives considerable progenitor expansion and bone marrow (BM) remodelling. Single cell assays using Lin-Sca1+c-Kit+CD48-CD150+ long-term hematopoietic stem cells (LT-HSCs) revealed elevated LT-HSC activation and proliferation in vitro. However, the increased activation did not come at the expense of LT-HSC functionality as measured by in vivo serial transplantations. ITP progression was associated with considerable BM vasodilation and angiogenesis, as well as a 2-fold increase in local production of CXCL12; a cytokine essential for LT-HSC function and BM homing expressed at high levels by LepR+ BM stromal cells. This was associated with a 1.5-fold increase in LepR+ BM stromal cells and a 5.5-fold improvement in progenitor homing to the BM. Whereas the increase in stromal cells was transient and reverted back to baseline after platelet count returned to normal, vasculature changes in the BM persisted. Together, these studies demonstrate that LT-HSCs expand in response to ITP, and that LT-HSC functionality during sustained hematopoietic stress is maintained through an adapting BM microenvironment.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3856-3856
Author(s):  
Renata Dmitrieva ◽  
Izida Minullina ◽  
Anna Bilibina ◽  
Olga Tarasova ◽  
Sergey Anisimov ◽  
...  

Abstract Abstract 3856 Cell therapy with MSCs appears to be a promising method for treatment of different disorders. Bone marrow and fat tissue are both considered as a prospective source of MSCs for therapeutic applications. However, the differences in functional properties of specific populations of MSC derived from these two tissues of the same patient are still poorly investigated. Patients and methods: 23 cardiovasular patients and 4 healthy donors were involved in this study. All patients were enrolled in programme funded by EU FP7. MSC cultures from BM (BM-MSC) and subcutaneous adipose (F-MSC) of the same patient were evaluated in successive passages for immunophenotype (FACS analysis), frequency of colony-forming units (CFU) in MSC population, frequency of adipo- and osteo-progenitors (CFU-Ad, CFU-Ost) in the same population and for dynamic of changes in these properties with successive passage. CFU, CFU-Ad and CFU-Ost were studied by limiting dilution assay followed by induction of adipo- or osteo- differentiation as described (Mitchell et al. Stem Cells 2006) with some modifications. Cell suspension was serially diluted two folds across the 8 columns of 96-well plates, resulting in columns containing from 50 to 0,39 cells per well. After 10 days of culture the number of positive and negative wells was determined for each cell concentration and CFU frequency was calculated. Then plates were induced to undergo adipogenesis and osteogenesis and CFU-Ad was determined by Oil Red staining and CFU-Ost by Alizarin Red staining after 14 and 21 days respectively. All calculations were performed as for CFU. Results. Significant difference was observed in immunophenotype of MSC derived from different tissues of the same donor: while both BM- and F-MSC were positive for stromal cell-associated markers CD105, CD90 and CD73 and were negative for hematopoietic lineage cells markers CD34, CD19, CD14, CD45, the population of CD146+ cells was more abundant in BM-MSC than in F-MSC at passage 1 (39,6%± 8,3% vs 5,6 %± 1,8%; p<0,005), this population of cells further declined in successive passages by passage 5 (17,7%± 6,9% in BM-MSC vs 1,5%± 0,65% in F-MSC; p<0,01); (Figure 1). To further characterize MSC derived from BM and F we studied the frequency of CFU at passages P1-P4, and CFU-Ad and CFU-Ost at passages 2 and 4. Results are shown in Tables 1 and 2. These data support the idea that in vitro expanded BM- and F- derived MSC differ in their properties. While the frequency of CFU in BM derived MSC population declined by as much as 4-fold by passage 4, there was 2-fold increase in frequency of CFU in Ad-MSC over the same period of culturing and this frequency remain unchanged up to passage 7 (data not shown). These data are consistent with our observation that cultures of BM-MSC showed first signs of senescence (senescence-associated β-galactosidase activity) as early as at passage 4,3±0.5 while the first signs of senescence in Ad-MSC cultures were seen at passage 6,67±0,66 (p<0,02). We assume that early decline in CFU frequency in BM-MSC in vitro is because of cell senescence. Importantly, the difference in CFU-Ad and CFU-Ost was discovered: frequency of both declined significantly in BM-MSC by passage 4, while frequency of Ad- and Ost- lineage progenitors remain unchanged in F-MSC cultures. Conclusion: For a first time a broad study was performed to compare MSC derived from BM and adipose tissues for immunophenotype, self-renewal and differentiation potential. We have found that BM-MSC and F-MSC differ significantly in all these characteristics. Differences in BM- and F-MSC could be explained by their ontogeny and/or different microenvironment in “parent” tissue. These variations could affect their efficacy in different therapeutic applications. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document