GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Blood Advances, American Society of Hematology, Vol. 5, No. 21 ( 2021-11-09), p. 4380-4392
    Abstract: Homing and engraftment of hematopoietic stem/progenitor cells (HSPCs) into the bone marrow (BM) microenvironment are tightly regulated by the chemokine stromal cell–derived factor-1 (SDF-1) and its G-protein–coupled receptor C-X-C motif chemokine receptor 4 (CXCR4), which on engagement with G-protein subunits, trigger downstream migratory signals. Regulators of G-protein signaling (RGS) are GTPase-accelerating protein of the Gα subunit and R4 subfamily members have been implicated in SDF-1–directed trafficking of mature hematopoietic cells, yet their expression and influence on HSPCs remain mostly unknown. Here, we demonstrated that human CD34+ cells expressed multiple R4 RGS genes, of which RGS1, RGS2, RGS13, and RGS16 were significantly upregulated by SDF-1 in a CXCR4-dependent fashion. Forced overexpression of RGS1, RGS13, or RGS16 in CD34+ cells not only inhibited SDF-1–directed migration, calcium mobilization, and phosphorylation of AKT, ERK, and STAT3 in vitro, but also markedly reduced BM engraftment in transplanted NOD/SCID mice. Genome-wide microarray analysis of RGS-overexpressing CD34+ cells detected downregulation of multiple effectors with established roles in stem cell trafficking/maintenance. Convincingly, gain-of-function of selected effectors or ex vivo priming with their ligands significantly enhanced HSPC engraftment. We also constructed an evidence-based network illustrating the overlapping mechanisms of RGS1, RGS13, and RGS16 downstream of SDF-1/CXCR4 and Gαi. This model shows that these RGS members mediate compromised kinase signaling and negative regulation of stem cell functions, complement activation, proteolysis, and cell migration. Collectively, this study uncovers an essential inhibitory role of specific R4 RGS proteins in stem cell engraftment, which could potentially be exploited to develop improved clinical HSPC transplantation protocols.
    Type of Medium: Online Resource
    ISSN: 2473-9529 , 2473-9537
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2021
    detail.hit.zdb_id: 2876449-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 1169-1169
    Abstract: Migration, homing and engraftment of hematopoietic stem/progenitor cells depend critically on the SDF-1/CXCR4 axis. We previously identified the tetraspanin CD9 as a downstream signal of this axis, and it regulates short-term homing of cord blood (CB) CD34+ cells (Leung et al, Blood, 2011). However, its roles in stem cell engraftment, mobilization and the underlying mechanisms have not been described. Here, we provided evidence that CD9 blockade profoundly reduced long-term bone marrow (BM; 70.9% inhibition; P = .0089) and splenic engraftment (87.8% inhibition; P = .0179) of CB CD34+ cells (n = 6) in the NOD/SCID mouse xenotransplantation model, without biasing specific lineage commitment. Interestingly, significant increase in the CD34+CD9+ subsets were observed in the BM (9.6-fold; P 〈 .0001) and spleens (9.8-fold; P = .0014) of engrafted animals (n = 3-4), indicating that CD9 expression on CD34+ cells is up-regulated during engraftment in the SDF-1-rich hematopoietic niches. Analysis of paired BM and peripheral blood (PB) samples from healthy donors revealed higher CD9 expressions in BM-resident CD34+ cells (46.0% CD9+ cells in BM vs 26.5% in PB; n = 13, P = .0035). Consistently, CD34+ cells in granulocyte colony-stimulating factor (G-CSF)-mobilized peripheral blood (MPB) expressed lower levels of CD9 (32.3% CD9+ cells; n = 25), when compared with those in BM (47.7% CD9+ cells; n = 16, P = .0030). In vitro exposure of MPB CD34+ cells to SDF-1 significantly enhanced CD9 expression (1.5-fold increase; n = 4, P = .0060). Treatment of NOD/SCID chimeric mice with G-CSF decreased the CD34+CD9+ subsets in the BM from 79.2% to 62.4% (n = 8, P = .0179). These data indicate that CD9 expression is down-regulated during egress or mobilization of CD34+ cells. To investigate the possible mechanisms, we performed a VCAM-1 (counter receptor of the VLA-4 integrin) binding assay on BM CD34+ cells. Our results demonstrated that CD34+CD9+ cells preferentially bound to soluble VCAM-1 (17.2%-51.4% VCAM-1-bound cells in CD9+ cells vs 12.8%-25.9% in CD9- cells; n = 10, P ≤ .0003), suggesting that CD9+ cells possess higher VLA-4 activity. Concomitant with decreased CD9 expression, MPB CD34+ cells exhibited lower VCAM-1 binding ability (2.8%-4.0% VCAM-1-bound cells; n = 3), when compared to BM CD34+ cells (15.5%-37.7%; n = 10, P 〈 .0130). In vivo treatment of NOD/SCID chimeric mice with G-CSF reduced VCAM-1 binding of CD34+ cells in the BM by 49.0% (n = 5, P = .0010). Importantly, overexpression of CD9 in CB CD34+ cells promoted VCAM-1 binding by 39.5% (n = 3, P = .0391), thus providing evidence that CD9 regulates VLA-4 activity. Preliminary results also indicated that enforcing CD9 expression in CB CD34+ cells could enhance their homing and engraftment in the NOD/SCID mouse model. Our findings collectively established that CD9 expression and associated integrin VLA-4 activity are dynamically regulated in the BM microenvironment, which may represent important events in governing stem cell engraftment and mobilization. Strategies to modify CD9 expression could be developed to enhance engraftment or mobilization of CD34+ cells. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 2371-2371
    Abstract: The stromal cell-derived factor-1 (SDF-1)/chemokine C-X-C receptor 4 (CXCR4) axis is crucial to migration and homing of hematopoietic stem/progenitor cells (HSC). In a previous study, we reported the expression profile of human cord blood CD34+ cells in response to a short-term exposure of SDF-1 (Leung et al, Blood, 2011). We further identified that expression levels of several R4 Regulator of G-protein signaling (RGS) family proteins were upregulated upon SDF-1 treatment. RGS family proteins are known to negatively control G-protein-coupled receptor signal transduction through their GTPase-accelerating activity. In several types of hematopoietic cells (e.g., B lymphocytes), responses to SDF-1 are subjected to regulation by RGS proteins. However, their expression patterns and functional roles in HSC are poorly characterized. We first assessed the basal expressions of 10 R4 RGS proteins by quantitative RT-PCR. Our results demonstrated that cord blood CD34+ cells expressed relatively high mRNA level of RGS1, RGS2 and RGS3, and moderate level of RGS5, RGS13, RGS16 and RGS18 (n = 3). Expression of RGS8 was barely detectable, while RGS4 and RGS21 expression was not detectable. A short-term exposure (1-24 hours) of CD34+ cells to SDF-1 (100 ng/mL, n = 4) significantly increased expressions of RGS1 (1.5 fold, P = .002), RGS13 (1.8 fold, P = .044), and to a lesser extent RGS2 and RGS3 (both 1.3 fold; P 〈 .032). Expressions of RGS5, RGS16 and RGS18 were not affected by SDF-1 stimulation. Preincubation of CD34+ cells with the CXCR4 antagonist AMD3100 resulted in 82.2% and 56.2% inhibition of SDF-1-induced RGS1 and RGS13 expressions respectively. To investigate the functional roles of RGS1 and RGS13 in SDF-1-mediated responses, we introduced GFP-tagged cDNA clones into cord blood CD34+ cells by lentiviral transduction. Using the spleen focus-forming virus (SFFV) promoter, we achieved 55.2% transduction efficiency in control GFP vector-transduced cells, 39.4% in RGS1-transduced cells and 51.7% in RGS13-transduced cells. Quantitative RT-PCR further confirmed RGS1 (76 fold) and RGS13 (3,641 fold) overexpression in transduced CD34 cells. We showed that SDF-1-directed migration was inhibited by 18.5% in RGS1-transduced cells, but not in RGS13-transduced cells. In RGS1-transduced cells, significantly less GFP+ cells was found in the migrating fraction (51% in non-migrating vs 28% in migrating fraction). We further demonstrated that actin polymerization, an early response to SDF-1 stimulus, was reduced by 9.5% in RGS13-transduced cells but not in RGS1-transduced cells. In the NOD/SCID mouse xenotransplantation model, preliminary results showed that bone marrow homing of CD34+ cells was impaired in RGS1-overexpressing (54.4% reduction) or RGS13-overexpressing (28.1% reduction) cells, when compared to control GFP-transduced cells. Splenic homing of CD34+ cells was not affected by either RGS1 or RGS13. Taken together, we provided the first expression profile of R4 RGS proteins in cord blood CD34+ cells, and their expressional changes in response to SDF-1 stimulation. We also provided evidence that RGS1 and RGS13 regulate specific SDF-1-mediated responses and, importantly, both negatively affect homing of CD34+ cells. Strategies to inhibit RGS1 or RGS13 signaling could be a potential method for enhancing HSC homing and their long-term engraftment for immunologic reconstitution. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Leukemia, Springer Science and Business Media LLC, Vol. 34, No. 3 ( 2020-03), p. 709-720
    Type of Medium: Online Resource
    ISSN: 0887-6924 , 1476-5551
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2008023-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 995-995
    Abstract: Gram-negative bacterial infection is a serious condition in neonates which could lead to septicemic shock, disseminated intravascular coagulation and death. LPS, the cell wall component of Gram-negative bacteria is a potent stimulator of the host immune response system, mediated by binding to toll-like receptor (TLR)-4 and CD14 receptors. Neutrophils are the first line of innate defense which possess antimicrobial and acute inflammatory activities. Neonatal neutrophils, though incompletely characterized, are suggested to be functionally immature and compromised in their bactericidal capacity compared with adults. We performed a comparative genome-wide expression array analysis on purified ( 〉 95%) human cord blood (term delivery) neutrophils upon challenge by LPS for 4 hours at 100 ng/mL. With the criteria of 2-fold differences and statistical significance (P 〈 0.01), 1236 transcripts were increased and 1633 transcripts were decreased in LPS-treated neutrophils (n=3). A panel of 20 target genes was selected by their magnitude of regulation, novelty and relevant immune functions and validated by qPCR. Eleven of these genes could be linked upstream or downstream of the novel transcription factor, nuclear receptor subfamily-4, group-A, member-2 (NR4A2) by pathway analysis (Metacore). We thus proposed a regulatory network mediated by binding of LPS to TLR4/CD14 receptors, leading to NR4A2 upregulation, thereby promoting a cascade of downstream effector genes associated with antibacterial and/or inflammatory activities in neutrophils. To validate the upstream signal cascade of LPS-induced regulation of NR4A2, neonatal neutrophils were pretreated with specific TLR-4, p38MAPK or NF-κB inhibitors, and stimulated with LPS, followed by quantification of target gene expression by qPCR (n=6 in each group). Our results showed that LPS-induced NR4A2 expression was significantly decreased (all P 〈 0.05) after pretreatment with TLR-4 antibody (0.47 fold), p38MAPK inhibitor SB203580 (0.27 fold), and NF-κB inhibitors BAY-117082 (0.14 fold) and pyrrolidine dithiocarbamate (0.07 fold). These results suggested that NR4A2 could be upregulated by LPS-receptor-mediated p38MAPK and NF-κB signals. To compare the capacity and developmental regulation of LPS-induced responses of NR4A2-associated signals, purified cord blood neutrophils from preterm (gestational age 33-36 weeks) and term (37-40 weeks) neonates, and peripheral blood from healthy male adult (age 25-30 years old) were evaluated (n=10 in each group). LPS-induced expression of NR4A2 and associated molecular signals (EGR1, PPARG, NF-κB, ATOH8, G0S2, GPR84, IL-8, PTGS2, TNF-α, CAMK2G and GCH1) were measured by qPCR. Our result showed that expressions of these target genes were significantly up- or downregulated in LPS-activated neutrophils of neonates and adults. However, at the basal level, expressions of EGR1, GOS2, IL-8 and PTSG2 were significantly lower (P 〈 0.05) in preterm or term neonatal neutrophils compare with those in adults, showing that there might be compromised levels in neonates at the unstimulated status. Upon stimulation with LPS, expressions of GOS2 and IL-8 were significantly lower (P 〈 0.05) in preterm or term neonates than in adult, indicating that there could be developmental regulation of NR4A2 downstream genes in neutrophils secondary to LPS stimulations. Correlation analysis showed that the expressions of EGR1 and PTGS2 were positively correlated with NR4A2 in LPS-induced neutrophils from both term (EGR1: R=0.805, P 〈 0.05; PTGS2: R=0.912, P 〈 0.05) and preterm neonates (EGR1: R=0.636, P 〈 0.05; PTGS2: R=0.830, P 〈 0.05), but not in adult peripheral neutrophils. These results indicated that there could be developmental differences in the regulation of the NR4A2 immune cascade involving apoptosis (EGR1, GOS2)/inflammatory cytokine (IL-8) and prostaglandin synthesis (PTGS2) between adult and neonatal neutrophils at basal and/or LPS-activation states. In summary, we reported the expressional profile and deregulated target genes in neonatal neutrophils upon stimulation by LPS and identified the downstream NR4A2 immune cascade that could be regulated by LPS, p38MAPK and NF-κB signals, as well as the developmental status. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Society of Hematology ; 2021
    In:  Blood Vol. 138, No. Supplement 1 ( 2021-11-05), p. 2297-2297
    In: Blood, American Society of Hematology, Vol. 138, No. Supplement 1 ( 2021-11-05), p. 2297-2297
    Abstract: Background/Aims: Despite advances in chemotherapy-based treatment protocols, the outcomes of children with acute myeloid leukemia (AML) remain suboptimal. Implementation of targeted therapy based solely on genomics is challenging due to the complex mutational patterns and scarcity of pharmacologic agents for most lesions. In addition, pediatric and adult AML are genetically and biologically distinct, which poses a major hurdle for extrapolation of new agents approved for adult AML to the pediatric population. This study aims to adopt a functional approach that directly measure the response of patient-derived leukemic cells to targeted agents, and to establish the drug sensitivity pattern and identify candidates of immediate clinical relevance for precision usage in high-risk pediatric AML. Methods: A high-throughput drug screening, comprising 39 targeted agents (2 in Phase I, 10 in Phase II, 5 in Phase III, 22 FDA-approved) and 6 conventional chemotherapeutics, was performed on 30 pediatric AML samples collected at diagnosis or relapse using a serum-free, cytokine-supported culture system. A counter-screen of active drugs on cord blood hematopoietic stem cells was accomplished to reveal leukemia-selective activities. The robustness of the drug testing platform for predicting in vivo activities was validated in xenograft models. Genomic profiling was complementarily performed to identify the genetic markers and underlying mechanisms of drug sensitivity. Patients with refractory AML were treated with targeted agents based on drug profiling results, and assessed for clinical responses. Results: Unsupervised clustering revealed 5 distinct clusters of drug response: highly active compounds (IC50 & lt;15 nM, 5 drugs); generally active compounds (IC50 & lt;250 nM, 11 drugs); compounds with bimodal activities (wide IC50 ranges, 3 drugs); generally inactive compounds (16 drugs); and inactive compounds (IC50 & gt;2000 nM, 10 drugs). Targeted agents, including Bcl-2, HDAC, proteasome, HSP and survivin inhibitors, had substantially higher potency and selectivity over standard chemotherapeutic agents. New agents approved for adult AML were essentially inactive in pediatric AML. Drug sensitivity ex vivo accurately predicted in vivo single-agent and combinatorial activities with cytarabine in cell line- and patient-derived xenografts. Targeted resequencing of a 141-gene panel revealed novel mutations of prognostic relevance, such as KMT2C, in pediatric AML and their vulnerability to targeted agents. Whole-genome RNA-sequencing identified distinct gene expression signatures shaping the response to individual drugs. Administration of venetoclax to a child with refractory AML resulted in rapid blast clearance and achieved long-term remission. Complementary genomic profiling on serial specimens dictated the dynamic drug responses during disease evolution. Conclusions: Our study establishes a reliable drug testing platform and a pediatric-specific drug response profile of AML, which enables an evidence-based selection of targeted agents for patients without treatment options and endows therapies increasingly precise and personalized. The study also generates a valuable gene-drug-clinical dataset that could be leveraged to address the fundamental and translational biology of pediatric AML. It will ultimately impact the future design of clinical trials and protocols for managing this life-threatening malignancy. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2021
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 5048-5048
    Abstract: RGS family proteins are known to negatively regulate G-protein-coupled receptor signaling through their GTPase-accelerating activity. In several types of hematopoietic cells (e.g., B lymphocytes and megakaryocytes), responses to stromal cell-derived factor-1 (SDF-1) are subjected to regulation by R4 subfamily RGS proteins. However, their expression patterns and functional roles in hematopoietic stem and progenitor cells (HSC) are poorly characterized. Here, we showed that human CD34+ HSC derived from cord blood (CB, n = 10) expressed 7 out of 10 R4 RGS proteins at mRNA level (RGS1-3, 5, 13, 16 and 18), whereas expressions of RGS4, 8 and 21 were undetectable. Exposure of CB CD34+ cells to SDF-1 significantly increased RGS1, 2, 13 and 16 expressions and decreased RGS3 and 18 expressions (P ≤ 0.0402, n = 5). Expressions of RGS1, 13 and 16 were significantly higher in bone marrow (BM, n = 10) CD34+ cells when compared to mobilized peripheral blood (MPB, n = 5) CD34+ cells (P ≤ 0.0160), while RGS3 and 18 expressions were lower in BM CD34+ cells (P ≤ 0.0471), suggesting a SDF-1- and niche-dependent regulation of RGS expressions. To investigate the potential involvement of RGS proteins in SDF-1-mediated homing-related functions, we introduced RGS overexpression constructs into CB CD34+ cells by lentiviral transduction. With 〉 80% transduction efficiency, we showed that overexpression of RGS1, 13 and 16 but not RGS2 significantly inhibited migration of CD34+ cells to a SDF-1 gradient (P ≤ 0.0391, n = 4-5). Similarly, RGS1, 13 and 16 overexpression suppressed SDF-1-induced Akt phosphorylation (n = 2), but none of them affected SDF-1-mediated actin polymerization (n = 3). In the NOD/SCID mouse xenotransplantation model, preliminary results showed that bone marrow homing was impaired in RGS1- (16.3% reduction), RGS13- (12.7% reduction) or RGS16-overexpressing CD34+ cells (33.7% reduction). Taken together, we provided the first evidence that expressions of R4 RGS proteins are regulated by the SDF-1/CXCR4 axis in human CD34+ HSC. We also presented evidence that specific R4 RGS proteins (RGS1, 13 and 16) negatively regulate in vitro SDF-1-mediated responses and in vivo homing of CD34+ cells, suggesting that RGS proteins may serve as a feedback mechanism to regulate SDF-1/CXCR4 signaling. Strategies to inhibit RGS signaling could thus be a potential method for enhancing efficiency of HSC homing and long-term engraftment, which is particularly important in the setting of CB transplantation. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Journal of Critical Care, Elsevier BV, Vol. 38 ( 2017-04), p. 57-61
    Type of Medium: Online Resource
    ISSN: 0883-9441
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2017
    detail.hit.zdb_id: 2041640-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 2906-2906
    Abstract: Background:In postnatal life, hematopoietic stem/progenitor cells (HSC) mostly reside in specialized bone marrow niches, however a low number of HSC is constantly released into the peripheral circulation. Emerging evidence suggests that these circulating HSC may play an active role in the host defense mechanism against bacterial infection by fostering the local production of tissue-resident innate immune cells. In adult patients with sepsis, CD34+ cells were significantly elevated in their circulation, which correlated with monocyte production and survival. Due to the immaturity of the immune system, preterm infants are vulnerable to life-threatening systemic infection. Although neonatal blood contains relatively high levels of CD34+ cells, there has been no knowledge on whether the same regulation of circulating CD34+ cells occurs in preterm infants in response to systemic infection, as in adults. Methods: We collected peripheral blood from preterm, very low birth weight ( 〈 1500 g) infants with suspected late-onset ( 〉 72 hours of age) infection requiring full sepsis evaluation and antibiotic treatment. Circulating CD34+ cells were assessed using 3-color flow cytometry (CD34, CD45 and 7-AAD), and enumerated using the ISHAGE gating strategy. Results: Of 39 episodes of suspected clinical sepsis investigated, 12 were blood culture-confirmed sepsis and 27 were non-infected episodes. There were no significant differences between infected and non-infected infants in gestational age, birth weight, postnatal age at the time of sepsis screening, Apgar scores at 1 and 5 minutes, male/female ratio or white cell count. However, there was a significant decrease in the frequency of CD34+ cells (percentage of total CD45+ cells) in infected infants [median (interquartile range), 0.08 (0.06-0.16) %], when compared to non-infected episodes [0.19 (0.12-0.34) %, P = 0.001] . Concomitant increase in the frequency of neutrophils [69 (59-75) % vs. 45 (34-53) %, P 〈 0.001] and decrease in lymphocytes [18 (13-22) vs. 32 (27-41) %, P 〈 0.001] were observed during these infection episodes. In addition, an inverse correlation was found between the frequencies of CD34+ cells and neutrophils (r = -0.471, P = 0.004), whereas a positive correlation was observed between the frequencies of CD34+ cells and lymphocytes (r = 0.485, P = 0.003). Interestingly, a rebound of CD34+ cells was evidenced at 24 hours after the commencement of antibiotic treatment in the sepsis group [0.12 (0.07-0.20) %, P = 0.012] but not in the non-infected group [0.21 (0.16-0.27) %, P= 0.847], suggesting that the level of circulating CD34+ cells may be under active control and correlate with clinical progress. Conclusions: This is the first evaluation of circulating CD34+ cells in preterm infants during an episode of systemic infection. Our data revealed a drop in the frequency of CD34+ cells in these infants at the onset of sepsis, which may be linked to the preferential commitment to neutrophilic lineage at the expense of lymphocyte production. Moreover, there is an opposite direction in the regulation of CD34+ cells in preterm infants when compared with adults affected by sepsis or other inflammatory conditions. This phenomenon may be attributed by defective mobilization, lower plasma levels of stem cell-mobilizing cytokines or insufficient stem cell pool in the bone marrow, which warrants further investigation. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 3237-3237
    Abstract: The stromal cell-derived factor-1 (SDF-1)/chemokine C-X-C receptor 4 (CXCR4) axis plays a critical role in homing, engraftment and retention of hematopoietic stem/progenitor cells. We previously demonstrated that expression of CD9 is a downstream signal of the SDF-1/CXCR4 axis, and that CD9 regulates short-term (20 hours) homing of cord blood (CB) CD34+ cells in the NOD/SCID mouse xenotransplantation model (Leung et al, Blood, 2011). Here, we provided further evidence that pretreatment of CB CD34+ cells with a CD9-neutralizing antibody significantly reduced their long-term (6 weeks) engraftment, as indicated by the presence of human CD45+ cells, in the recipient bone marrow and spleen by 70.9% (P = .0089) and 87.8% (P = .0179), respectively (n = 6). However, CD9 blockade did not bias specific lineage commitment, including the CD14+ monocytic, CD33+ myeloid, CD19+ B-lymphoid and CD34+ stem/progenitor cells (n = 4). We also observed an increase of the CD34+CD9+ subsets in the bone marrow (9.6-fold; P 〈 .0001) and spleens (9.8-fold; P = .0014) of engrafted animals (n = 3-4). These data indicate that CD9 possesses important functions in regulating stem cell engraftment and its expression level on CD34+ cells is up-regulated in the target hematopoietic organs. Analysis of paired bone marrow (BM) and peripheral blood (PB) samples from healthy donors revealed a higher CD9 expression in BM-resident CD34+ cells (57.3% ± 8.1% CD9+ cells in BM vs. 29.3% ± 5.8% in PB; n = 5, P = 0.0478). Consistently, CD34+ cells in granulocyte colony-stimulating factor (G-CSF)-mobilized peripheral blood (MPB) expressed lower levels of CD9 (33.8% ± 3.0% CD9+ cells, n = 24), when compared with those in BM (56.4% ± 4.9% CD9+ cells, n = 8, P = 0.0025). In vitro exposure of MPB CD34+ cells to SDF-1 significantly enhanced CD9 expression (1.55-fold increase, n = 4, P = 0.0103), concomitant with a 75.2% reduction in the CD34+CXCR4+ subsets (P = 0.0118). Treatment of NOD/SCID chimeric mice with G-CSF increased the frequency of circulating CD45+ cells (3.4-fold) and CD34+ cells (3.3-fold), and substantially decreased the CD34+CD9+ subsets in the BM from 75.8% to 30.8%. Importantly, the decline in CD9 levels during G-CSF mobilization was also observed in the CD34+CD38-/low primitive stem cell subpopulation. Interestingly, in vitro treatment of BM CD34+ cells with G-CSF did not affect CD9 expression (n = 3), suggesting that a signaling intermediate is required for G-CSF-mediated CD9 down-regulation in vivo. Transwell migration assay revealed a significant enrichment of CD9- cells that were migrated towards a SDF-1 gradient (n = 4 for BM CD34+ cells, P = 0.0074; n = 7 for CB CD34+ cells, P = 0.0258), implicating that CD9 might negatively regulate stem cell motility. In contrast, pretreatment with the CD9-neutralizing antibody inhibited adhesion of CD34+ cells to the osteoblastic cell line Saos-2 by 33.5% (n = 2). Our results collectively suggest a previously unrecognized role of CD9 in stem cell retention by dual regulation of cell motility and adhesion, and reveal a dynamic regulation of CD9 expression in the BM microenvironment, which might represent an important event in controlling stem cell homing and mobilization. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...