To see the other types of publications on this topic, follow the link: CD309.

Journal articles on the topic 'CD309'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'CD309.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Meamr, Rokhsareh, Hamidreza Nikyar, Ahmad Chitsaz, Leila Dehghani, and Maryam Nasri. "Evaluation of related variables on endothelial progenitor cells in first transient ischemic attack." Journal of Shahrekord University of Medical Sciences 21, no. 4 (August 30, 2019): 157–62. http://dx.doi.org/10.34172/jsums.2019.28.

Full text
Abstract:
Background and aims: In a transient ischemic attack (TIA), the activation of the endothelial progenitor cells (EPCs) has a prominent role in the formation of collateral vessels. The aim of this study was to evaluate the role of effective variables (e.g., hypertension, diabetes, hyperlipidemia, cardiovascular diseases, a family history of cardiovascular diseases, and statin therapy on the level of EPCs in TIA. Methods: Thirty patients suffering from the first attack of TIA, without having a history of acute cerebral injury, surgery or trauma, and blood disorders were enrolled in this cross-sectional study. Then, flow cytometry was utilized to estimate the level of EPCs CD34 and CD309 and the results were evaluated based on the t test or the Mann-Whitney test. Finally, the Pearson or Spearman correlation was used to establish the relationship between the variables. The level of P<0.05 was considered statistically significant in this study. Results: The mean±SD number of CD309 in patients with hyperlipidemia was less than those with no hyperlipidemia (3.35±2.77 vs. 5.59±3.85, P=0.02) and diabetic patients had a significantly higher number of CD309 compared to non-diabetics (6.14±4.89 vs, 3.5± 3.49, P=0.05). Conversely, the mean number of CD34 in groups with or without the studied variables was not statistically significant. The results further revealed that the average total of CD309 and CD34 was significantly lower in patients with hyperlipidemia as compared to those with no sign of hyperlipidemia (9.44± 3.05 vs. 6.67±4.6, P=0.02). Using logistic regression, the intended variables demonstrated no significant effects on endothelial cells, and the relationship between age and the number of progenitor cells was not significant. Conclusion: In our study, only hyperlipidemia acted as a factor influencing the numbers of EPCs. Therefore, more studies with larger sample sizes are required to discover the role of these variables on progenitor cells in TIA.
APA, Harvard, Vancouver, ISO, and other styles
2

Huang, Zhi-Xin, Jin Fang, Chang-Hua Zhou, Jie Zeng, Dong Yang, and Zhenguo Liu. "CD34+ cells and endothelial progenitor cell subpopulations are associated with cerebral small vessel disease burden." Biomarkers in Medicine 15, no. 3 (February 2021): 191–200. http://dx.doi.org/10.2217/bmm-2020-0350.

Full text
Abstract:
Background: Endothelial dysfunction is considered to be involved in the pathogenesis of cerebral small vessel disease (CSVD). Endothelial progenitor cells are associated with endothelial dysfunction. The present study was designed to investigate the correlation between the populations of circulating CD34-positive cells and endothelial progenitor cells and CSVD burden. Methodology & results: A total of 364 patients with confirmed diagnosis of CSVD were included in this prospective study. Multiple ordinal logistic regression analyses showed that subjects with higher CSVD burden had significantly decreased circulating CD34+ cell level (odds ratio [OR], 0.42; p = 0.034) and significantly increased levels of circulating CD34+CD133+CD309+ and CD34+CD133+ cells (OR 1.07, p = 0.031; OR 1.03, p = 0.001, respectively), compared with patients with lower CSVD burden. Conclusion: The findings suggest that the levels of circulating CD34+ cells, CD34+CD133+CD309+ cells and CD34+CD133+ cells may be used as potential biomarkers to monitor the disease progression of CSVD.
APA, Harvard, Vancouver, ISO, and other styles
3

Younes, A., U. Consoli, V. Snell, K. Clodi, K. O. Kliche, J. L. Palmer, H. J. Gruss, et al. "CD30 ligand in lymphoma patients with CD30+ tumors." Journal of Clinical Oncology 15, no. 11 (November 1997): 3355–62. http://dx.doi.org/10.1200/jco.1997.15.11.3355.

Full text
Abstract:
PURPOSE CD30 ligand (CD30L), which is expressed on resting B and activated T lymphocytes, can induce cell death in several CD30+ cell lines. Patients with CD30+ tumors (Hodgkin's disease and Ki-1+ non-Hodgkin's lymphoma) frequently have elevated soluble CD30 (sCD30) levels in their serum, which correlates with a poor prognosis. The role of sCD30 in protecting tumor cells from CD30L-mediated cell death and the pattern of CD30L expression on human peripheral-blood lymphocytes (PBLs) of normal donors and patients with CD30+ tumors are investigated. MATERIALS AND METHODS CD30L surface protein expression was determined by two-color flow cytometry on PBLs of patients with CD30+ tumors and normal individuals. CD30L levels were determined on subsets of PBLs before and after stimulation with phytohemagglutinin (PHA), anti-CD3 antibody, or CD40L. sCD30 was measured by enzyme-linked immunosorbent assay (ELISA). The apoptotic activity of membrane-bound CD30L was tested in a CD30+ cell line by the annexin V-binding method. RESULTS Unstimulated T lymphocytes of normal donors and patients with lymphoma rarely expressed CD30L surface protein, but were able to express it after stimulation with PHA or anti-CD3 antibody. Resting B cells of patients with CD30+ tumors had lower levels of detectable surface CD30L compared with normal donors (mean, 55% and 80.6%, respectively; P = .0008). Patients with high levels of serum sCD30 had lower detectable levels of CD30L on their PBLs (R2 = .72, P = .0008) and exogenous sCD30 blocked membrane-bound CD30L-mediated apoptosis in a CD30+ cell line. CONCLUSION In patients with CD30+ tumors, sCD30 can decrease the availability of CD30L on PBLs. Blocking the apoptosis-inducing activity of CD30L by its soluble receptor may explain how CD30+ tumors escape immunosurveillance and may be related to the reported poor prognosis of patients who have elevated sCD30 levels.
APA, Harvard, Vancouver, ISO, and other styles
4

Berezin, Alexander E., Alexander A. Kremzer, Daniel Petrovich, Ioana Mozos, and Alexander A. Berezin. "Association of growth-differentiation factor-15 with the number of circulating proangiogenic endothelial progenitor cells in patients with type 2 diabetes mellitus." Biomedical Research and Therapy 5, no. 7 (July 27, 2018): 2480–92. http://dx.doi.org/10.15419/bmrat.v5i7.458.

Full text
Abstract:
The objective: to investigate the relationship between levels of growth differentiation factor-15 (GDF-15) and circulating number of endothelial progenitor cells (EPCs) with angiopoietin phenotypes: CD34+CD14+CD309+, and CD34+CD14+CD309+Tie2+ in patients with type 2 DM. Methods: The study was retrospectively involved 76 patients with type 2 DM aged 38 to 55 years and 30 healthy volunteers. Data collection included demographic and anthropometric information, hemodynamic performances and biomarkers of the diseases. Flow cytometry was used to determine EPCs' populations. Results: The levels of GDF-15 in peripheral blood of diabetics associated with age (r = 0.31, P = 0.044), high-sensitive C-reactive protein [hs-CRP] (r = 0.40, P = 0.001), smoking (r = 0.38, P = 0.001), body mass index [BMI] (r = 0.34, P = 0.001), LDL cholesterol (r = 0.28, P = 0.001), glycated hemoglobin [HbA1c] (r = -0.28, P = 0.001), number of CV risk factors (r = 0.26, P = 0.001). In univariate logistic regression analysis we found that level of GDF-15 ≥ 618 pg/mL, hs-CRP ≥7.12 mg/L, HbA1c ≥6.4%, fasting glucose ≥6.7 mmol/L, and BMI ≥27.3 kg/m2 predicted deficiency of both angiopoetic phenotypes of EPCs. In multivariate logistic regression model GDF-15 ≥618 pg/mL demonstrated the best odds ratio values for declining of EPCs in diabetics in comparison with other predictors including BMI, HbA1c and hs-CRP. Conclusion: GDF-15 was remarkably evaluated in type 2 DM population to healthy volunteers, and it was an independent factor that contributes to mobilization and probably proliferation of endothelial precursors with high angiopoetic activity.
APA, Harvard, Vancouver, ISO, and other styles
5

Mori, M., C. Manuelli, N. Pimpinelli, C. Mavilia, E. Maggi, M. Santucci, B. Bianchi, P. Cappugi, B. Giannotti, and M. E. Kadin. "CD30-CD30 Ligand Interaction in Primary Cutaneous CD30+T-Cell Lymphomas: A Clue to the Pathophysiology of Clinical Regression." Blood 94, no. 9 (November 1, 1999): 3077–83. http://dx.doi.org/10.1182/blood.v94.9.3077.

Full text
Abstract:
Abstract Primary CD30+ cutaneous T-cell lymphomas (CTCLs) represent a spectrum of non-Hodgkin’s lymphomas (NHLs) that have been well defined at the clinical, histologic, and immunologic level. This group, which includes 2 main entities (large cell lymphoma and lymphomatoid papulosis [LyP]) and borderline cases, is characterized by the expression of CD30 antigen by neoplastic large cells at presentation, possible spontaneous regression of the skin lesions, and generally favorable clinical course. Although the functional relevance of CD30 and its natural ligand (CD30L) expression in most cases of NHL is presently undefined, previous studies indicate that CD30L is likely to mediate reduction of proliferation in CD30+ anaplastic large-cell NHL. No information is currently available concerning the expression of CD30L in primary CD30+ CTCLs. In this study, we investigated the immunophenotypic and genotypic expression of CD30 and CD30L in different developmental phases of skin lesions (growing v spontaneously regressing). By immunohistochemistry, CD30L expression was detected in regressing lesions only; by molecular analysis, the expression of CD30L was clearly higher in regressing lesions than in growing ones. CD30L, while expressed by some small lymphocytes, was most often coexpressed by CD30+neoplastic large cells, as demonstrated by 2-color immunofluorescence and by immunohistochemistry on paraffin sections. Taken together, these data suggest that CD30-CD30L interaction may play a role in the pathobiology of primary cutaneous CD30+lymphoproliferative disorders. In particular, CD30L (over)expression might have a major role in the mechanism of self-regression of skin lesions, the most distinctive clinical feature of this cutaneous lymphoma subtype.
APA, Harvard, Vancouver, ISO, and other styles
6

Mori, M., C. Manuelli, N. Pimpinelli, C. Mavilia, E. Maggi, M. Santucci, B. Bianchi, P. Cappugi, B. Giannotti, and M. E. Kadin. "CD30-CD30 Ligand Interaction in Primary Cutaneous CD30+T-Cell Lymphomas: A Clue to the Pathophysiology of Clinical Regression." Blood 94, no. 9 (November 1, 1999): 3077–83. http://dx.doi.org/10.1182/blood.v94.9.3077.421k28_3077_3083.

Full text
Abstract:
Primary CD30+ cutaneous T-cell lymphomas (CTCLs) represent a spectrum of non-Hodgkin’s lymphomas (NHLs) that have been well defined at the clinical, histologic, and immunologic level. This group, which includes 2 main entities (large cell lymphoma and lymphomatoid papulosis [LyP]) and borderline cases, is characterized by the expression of CD30 antigen by neoplastic large cells at presentation, possible spontaneous regression of the skin lesions, and generally favorable clinical course. Although the functional relevance of CD30 and its natural ligand (CD30L) expression in most cases of NHL is presently undefined, previous studies indicate that CD30L is likely to mediate reduction of proliferation in CD30+ anaplastic large-cell NHL. No information is currently available concerning the expression of CD30L in primary CD30+ CTCLs. In this study, we investigated the immunophenotypic and genotypic expression of CD30 and CD30L in different developmental phases of skin lesions (growing v spontaneously regressing). By immunohistochemistry, CD30L expression was detected in regressing lesions only; by molecular analysis, the expression of CD30L was clearly higher in regressing lesions than in growing ones. CD30L, while expressed by some small lymphocytes, was most often coexpressed by CD30+neoplastic large cells, as demonstrated by 2-color immunofluorescence and by immunohistochemistry on paraffin sections. Taken together, these data suggest that CD30-CD30L interaction may play a role in the pathobiology of primary cutaneous CD30+lymphoproliferative disorders. In particular, CD30L (over)expression might have a major role in the mechanism of self-regression of skin lesions, the most distinctive clinical feature of this cutaneous lymphoma subtype.
APA, Harvard, Vancouver, ISO, and other styles
7

Barbieri, Alessandro, Marzia Dolcino, Elisa Tinazzi, Antonella Rigo, Giuseppe Argentino, Giuseppe Patuzzo, Andrea Ottria, Ruggero Beri, Antonio Puccetti, and Claudio Lunardi. "Characterization of CD30/CD30L+Cells in Peripheral Blood and Synovial Fluid of Patients with Rheumatoid Arthritis." Journal of Immunology Research 2015 (2015): 1–10. http://dx.doi.org/10.1155/2015/729654.

Full text
Abstract:
The CD30/CD30L signalling system has been implicated in the pathogenesis of several autoimmune and inflammatory conditions. In rheumatoid arthritis (RA), soluble CD30 (sCD30) levels reflect the recruitment of CD30+T cells into the inflamed joints and correlate with a positive response to immunosuppressive therapy. The aim of our report was to clarify the role of CD30/CD30L signalling system in the pathogenesis of RA. Our analysis of the CD30L+T cell subsets in peripheral blood (PB) and synovial fluid (SF) of RA patients and of the related cytokine profiles suggests the involvement of CD30/CD30L signalling in polarization of T cells towards a Th17 phenotype with proinflammatory features. Moreover, in RA SF nearly 50% of Treg cells express CD30, probably as an attempt to downmodulate the ongoing inflammation. We also show here that the engagement of CD30L on neutrophils stimulated with CD30/Fc chimera may play a crucial role in RA inflammation since activated neutrophils release IL-8, thus potentially amplifying the local inflammatory damage. In conclusion, the results obtained suggest that the complex CD30/CD30L signalling pathway is implicated in the pathogenesis and progression of RA synovitis through a concerted action on several immune effector cells.
APA, Harvard, Vancouver, ISO, and other styles
8

Ryan, Maureen, Ryan Lyski, Lauren Bou, Ryan Heiser, Bryan Grogan, Dave Meyer, Steven Jin, et al. "SGN-CD30C, an Investigational CD30-Directed Camptothecin Antibody-Drug Conjugate (ADC), Shows Strong Anti Tumor Activity and Superior Tolerability in Preclinical Studies." Blood 136, Supplement 1 (November 5, 2020): 41–42. http://dx.doi.org/10.1182/blood-2020-136577.

Full text
Abstract:
SGN-CD30C, an investigational CD30-directed ADC, utilizes a potent novel camptothecin derivative as the cytotoxic payload. SGN-CD30C targets the same antigen as brentuximab vedotin (BV), though the payload has a different mechanism of action, namely inhibiting topoisomerase I rather than disrupting microtubules. Unlike monomethyl auristatin E, camptothecin-based therapies do not cause peripheral neuropathy clinically, suggesting that SGN-CD30C may have the potential to avoid one of the most common adverse events associated with BV. In preclinical studies, SGN-CD30C demonstrated strong monotherapy activity, inducing durable tumor regressions in multiple lymphoma models and eliciting cures in a BV-resistance tumor model (Figure 1). Moreover, SGN-CD30C exhibits many of the desirable attributes associated with BV, notably, bystander activity and CD30+ T regulatory cell (Treg) depletion. Using an admixed model of CD30+ and CD30-tumor cells, SGN-CD30C demonstrated robust anti-tumor activity in vivo, confirming SGN-CD30C can induce bystander killing of antigen-negative tumor cells in CD30-heterogeneous tumors. Additionally, SGN-CD30C depleted CD30+ Treg cells in vitro (Figure 2), suggesting it has the potential to exert activity through multiple mechanisms of action. SGN-CD30C was well-tolerated in non-human primate toxicology studies and demonstrated ~6-fold higher maximum tolerated dose when compared to BV. The primary toxicity for SGN-CD30C in non-human primates (NHP) studies was anemia due primarily to bone marrow suppression of erythropoiesis. SGN-CD30C had no effect on neutrophil counts and caused only minimal to mild decreases in platelets. The lack of significant neutropenia seen with SGN-CD30C contrasts with BV, indicating the two ADCs may have non-overlapping dose limiting toxicities. Hematology parameters show SGN-CD30C is tolerated at a 10-fold higher dose than BV with weekly dosing, suggesting SGN-CD30C may offer the potential for increased dose density. In summary, our data show SGN-CD30C is a compelling therapeutic candidate for CD30-positive malignancies. The distinct mechanism of action, strong anti-tumor activity, and enhanced tolerability provide a strong rationale to clinically investigate SGN-CD30C across the CD30-expressing lymphoma landscape. A Phase 1 clinical trial is planned to investigate SGN-CD30C in relapsed and refractory lymphoma. Disclosures Heiser: Seattle Genetics: Current Employment, Current equity holder in publicly-traded company. Grogan:Seattle Genetics: Current Employment, Current equity holder in publicly-traded company. Jin:Seattle Genetics: Current Employment, Current equity holder in publicly-traded company. Conerly:Seattle Genetics: Current Employment, Current equity holder in publicly-traded company.
APA, Harvard, Vancouver, ISO, and other styles
9

Bratseth, Vibeke, Hanna D. Margeirsdottir, Gemma Chiva-Blanch, Martin Heier, Svein Solheim, Harald Arnesen, Knut Dahl-Jørgensen, and Ingebjørg Seljeflot. "Annexin V+ Microvesicles in Children and Adolescents with Type 1 Diabetes: A Prospective Cohort Study." Journal of Diabetes Research 2020 (March 30, 2020): 1–8. http://dx.doi.org/10.1155/2020/7216863.

Full text
Abstract:
Background. Type 1 diabetes is a chronic disease including hyperglycemia and accelerated atherosclerosis, with high risk of micro- and macrovascular complications. Circulating microvesicles (cMVs) are procoagulant cell fragments shed during activation/apoptosis and discussed to be markers of vascular dysfunction and hypercoagulability. Limited knowledge exists on hypercoagulability in young diabetics. We aimed to investigate cMVs over a five-year period in children/adolescents with type 1 diabetes compared with controls and any associations with glycemic control and cardiovascular risk factors. We hypothesized increased shedding of cMVs in type 1 diabetes in response to vascular activation. Methods. The cohort included type 1 diabetics (n=40) and healthy controls (n=40), mean age 14 years (range 11) at inclusion, randomly selected from the Norwegian Atherosclerosis and Childhood Diabetes (ACD) study. Citrated plasma was prepared and stored at -80°C until cMV analysis by flow cytometry. Results. Comparable levels of Annexin V (AV+) cMVs were observed at inclusion. At five-year follow-up, total AV+ cMVs were significantly lower in subjects with type 1 diabetes compared with controls; however, no significant differences were observed after adjusting for covariates. In the type 1 diabetes group, the total AV+, tissue factor-expressing AV+/CD142+, neutrophil-derived AV+/CD15+ and AV+/CD45+/CD15+, and endothelial-derived AV+/CD309+ and CD309+/CD34+ cMVs were inversely correlated with HbA1c (r=‐0.437, r=‐0.515, r=‐0.575, r=‐0.529, r=‐0.416, and r=‐0.445, respectively; all p≤0.01), however, only at inclusion. No significant correlations with cardiovascular risk factors were observed. Conclusions. Children/adolescents with type 1 diabetes show similar levels of AV+ cMVs as healthy controls and limited associations with glucose control. This indicates that our young diabetics on intensive insulin treatment have preserved vascular homeostasis and absence of procoagulant cMVs.
APA, Harvard, Vancouver, ISO, and other styles
10

Simhadri, Vijaya Lakshmi, Hinrich P. Hansen, Venkateswara R. Simhadri, Katrin S. Reiners, Martina Bessler, Andreas Engert, and Elke Pogge von Strandmann. "A novel role for reciprocal CD30-CD30L signaling in the cross-talk between natural killer and dendritic cells." Biological Chemistry 393, no. 1-2 (January 1, 2012): 101–6. http://dx.doi.org/10.1515/bc-2011-213.

Full text
Abstract:
Abstract The interplay between dendritic cells (DCs) and natural killer (NK) cells directs adaptive immune responses. The molecular basis of the cross-talk is largely undefined. Here, we provide evidence for a contribution of CD30 (TNFRSF8) and its ligand CD30L (TNFSF8) expressed on NK cells and DCs, respectively. We demonstrate that CD30-mediated engagement of CD30L induced cytokine secretion from immature DCs via the mitogen-activated protein kinase pathway. Moreover, CD30L engagement promoted differentiation to mature DCs. On the contrary, the engagement of CD30 on NK cells resulted in an NF-κB-dependent release of TNF-α/IFN-γ. These data uncover a novel and unexpected role for CD30/CD30L that contributes to proinflammatory immune responses.
APA, Harvard, Vancouver, ISO, and other styles
11

Horie, R., K. Ito, M. Tatewaki, M. Nagai, S. Aizawa, M. Higashihara, T. Ishida, J. Inoue, H. Takizawa, and T. Watanabe. "A variant CD30 protein lacking extracellular and transmembrane domains is induced in HL-60 by tetradecanoylphorbol acetate and is expressed in alveolar macrophages." Blood 88, no. 7 (October 1, 1996): 2422–32. http://dx.doi.org/10.1182/blood.v88.7.2422.bloodjournal8872422.

Full text
Abstract:
We identified and cloned cDNAs for two novel CD30 mRNAs of 2.3 kb that are induced by 12-O-tetradecanoylphorbol 13-acetate (TPA) in the human myeloid leukemia cell line HL-60. These transcripts were transcribed from the intronic region just upstream of the exon coding for the transmembrane domain of the CD30 protein. The shorter cDNA had a deletion of 54 nucleotides corresponding to the 3′ region of the transmembrane domain of the CD30 and which was probably caused by alternative splicing. Translation of these transcripts appeared to start from the internal methionine codon at nucleotide position 289 that corresponds to that of 1612 in the CD30 cDNA, and encode a protein of 132 amino acid residues which corresponds exactly to the C-terminal cytoplasmic domain of CD30 protein. The calculated molecular mass of this variant CD30 (CD30v) protein was 14,087. Thus, the predicted CD30v protein retains most of the cytoplasmic region, but lacks the extracellular and transmembrane domains. Northern blots detected the expression of CD30v transcripts only in the lung and the TPA-stimulated HL-60 cell line. Translation of this mRNA in vitro produced a protein of 25 kD. Immunoblotting analysis with HCD30C1, a rabbit polyclonal antibody raised against the cytoplasmic domain of CD30 protein, detected proteins with an apparent Mr 25 kD expressed in TPA-stimulated HL-60 and COS-7 cells that were transfected with both types of CD30v cDNAs. Constitutive phosphorylation of the CD30v protein was demonstrated by in vitro labeling with [32P]. Immunohistochemical studies demonstrated CD30v protein was in alveolar macrophages. Cotransfection experiments using a kappa B-site-dependent reporter construct showed that CD30v can transactivate gene expression through activation of NF kappa B, as was noted on the authentic CD30 protein. Overexpression of the CD30v induced differentiation of HL-60 cells as evidenced by an increased NBT reduction activity. These observations provided new insights into the molecular heterogeneity and biological function of CD30 in myeloid cells.
APA, Harvard, Vancouver, ISO, and other styles
12

Romagnani, Paola, Francesco Annunziato, Roberto Manetti, Carmelo Mavilia, Laura Lasagni, Cinzia Manuelli, Gabriella B. Vannelli, et al. "High CD30 Ligand Expression by Epithelial Cells and Hassal's Corpuscles in the Medulla of Human Thymus." Blood 91, no. 9 (May 1, 1998): 3323–32. http://dx.doi.org/10.1182/blood.v91.9.3323.

Full text
Abstract:
Abstract CD30 is a member of tumor necrosis factor (TNF) receptor superfamily that is expressed by activated T cells in the presence of interleukin-4 (IL-4). Although CD30 can mediate a variety of signals, CD30-deficient mice have impaired negative selection of T cells, suggesting that at least in the context of murine thymus, CD30 is a cell death–mediating molecule. The ligand for CD30 (CD30L) is a membrane-associated glycoprotein related to TNF, which is known to be expressed mainly by activated T cells and other leukocytes. However, the nature of CD30L-expressing cells involved in the interaction with CD30+ thymocytes is unclear. We report here that in postnatal human thymus the great majority of CD30+ cells are double positive (CD4+CD8+), activated, IL-4 receptor–expressing T cells which selectively localize in the medullary areas. Moreover, many medullary epithelial cells and Hassal's corpuscles in the same thymus specimens showed unusually high expression of CD30L in comparison with other lymphoid or nonlymphoid tissues. These findings provide additional information on the nature and localization of CD30+ thymocytes and show that epithelial cells are the major holder of CD30L in the thymic medulla.
APA, Harvard, Vancouver, ISO, and other styles
13

Romagnani, Paola, Francesco Annunziato, Roberto Manetti, Carmelo Mavilia, Laura Lasagni, Cinzia Manuelli, Gabriella B. Vannelli, et al. "High CD30 Ligand Expression by Epithelial Cells and Hassal's Corpuscles in the Medulla of Human Thymus." Blood 91, no. 9 (May 1, 1998): 3323–32. http://dx.doi.org/10.1182/blood.v91.9.3323.3323_3323_3332.

Full text
Abstract:
CD30 is a member of tumor necrosis factor (TNF) receptor superfamily that is expressed by activated T cells in the presence of interleukin-4 (IL-4). Although CD30 can mediate a variety of signals, CD30-deficient mice have impaired negative selection of T cells, suggesting that at least in the context of murine thymus, CD30 is a cell death–mediating molecule. The ligand for CD30 (CD30L) is a membrane-associated glycoprotein related to TNF, which is known to be expressed mainly by activated T cells and other leukocytes. However, the nature of CD30L-expressing cells involved in the interaction with CD30+ thymocytes is unclear. We report here that in postnatal human thymus the great majority of CD30+ cells are double positive (CD4+CD8+), activated, IL-4 receptor–expressing T cells which selectively localize in the medullary areas. Moreover, many medullary epithelial cells and Hassal's corpuscles in the same thymus specimens showed unusually high expression of CD30L in comparison with other lymphoid or nonlymphoid tissues. These findings provide additional information on the nature and localization of CD30+ thymocytes and show that epithelial cells are the major holder of CD30L in the thymic medulla.
APA, Harvard, Vancouver, ISO, and other styles
14

Guo, Ying, Xun Sun, Kensuke Shibata, Hisakata Yamada, Hiromi Muta, Eckhard R. Podack, and Yasunobu Yoshikai. "CD30 Is Required for Activation of a Unique Subset of Interleukin-17A-Producing γδ T Cells in Innate Immunity against Mycobacterium bovis Bacillus Calmette-Guérin Infection." Infection and Immunity 81, no. 10 (August 5, 2013): 3923–34. http://dx.doi.org/10.1128/iai.00887-13.

Full text
Abstract:
ABSTRACTInterleukin-17A (IL-17A)-producing γδ T cells are known to be activated followingMycobacterium bovisbacillus Calmette-Guérin (BCG) infection. Here, we show that CD30, a member of the tumor necrosis factor (TNF) receptor superfamily, is important for activation of IL-17A-producing γδ T cells after BCG infection. Vγ1−Vγ4−γδ T cells preferentially expressing Vγ6/Vδ1 genes were identified as the major source of IL-17A in the peritoneal cavity during the early stage of BCG infection. The number of IL-17A-producing Vγ1−Vγ4−γδ T cells bearing Vγ6 increased in peritoneal exudate cells (PEC) of wild-type (WT) mice but not in those of CD30 knockout (KO) mice in response to BCG infection. Consistently, CD30 ligand (CD30L) or CD30 expression, predominantly by Vγ1−Vγ4−γδ T cells, was rapidly upregulated after BCG infection. Inhibition of CD30L/CD30 signaling byin vivoadministration of a soluble CD30 and immunoglobulin fusion protein (CD30-Ig) severely impaired activation of IL-17A-producing Vγ1−Vγ4−γδ T cells in WT mice, while stimulating CD30L/CD30 signaling byin vivoadministration of agonistic anti-CD30 monoclonal antibody (MAb) restored IL-17A production by Vγ1−Vγ4−γδ T cells in CD30L KO mice after BCG infection. These results suggest that CD30 signaling plays an important role in the activation of IL-17A-producing Vγ1−Vγ4−γδ T cells bearing Vγ6 at an early stage of BCG infection.
APA, Harvard, Vancouver, ISO, and other styles
15

Pinto, A., D. Aldinucci, A. Gloghini, V. Zagonel, M. Degan, S. Improta, S. Juzbasic, et al. "Human eosinophils express functional CD30 ligand and stimulate proliferation of a Hodgkin's disease cell line." Blood 88, no. 9 (November 1, 1996): 3299–305. http://dx.doi.org/10.1182/blood.v88.9.3299.bloodjournal8893299.

Full text
Abstract:
The presence of a prominent tissue eosinophilia represents a typical histopathologic hallmark of Hodgkin's disease (HD). To evaluate the putative role of eosinophils on tumor cell regulation in HD, we have analyzed these cells for the functional expression of CD30 ligand (CD30L), a surface molecule able to transduce CD30-mediated proliferation signals on Hodgkin's (H) and Reed-Sternberg (RS) cells. The results demonstrate that circulating and tissue eosinophils from normal donors and patients with HD or hypereosinophilic syndrome (HES), display CD30L mRNA and express CD30L protein, as shown by immunostaining with a specific monoclonal antibody (M80) and with a biotinylated soluble CD30-Fc fusion protein. The surface density of CD30L on eosinophils from HD and HES patients was remarkably higher compared with healthy donors, probably reflecting a cytokine-mediated upregulation in these pathologic conditions. Accordingly, we provide evidence that cytokines regulating eosinophils proliferation and activation, ie, interleukin-5 (IL-5), IL-3, and granulocyte-macrophage colony-stimulating factor (GM-CSF), are able to enhance the cellular density of CD30L on purified eosinophils from normal subjects. Finally, we show that native CD30L on human eosinophils is a functionally active surface structure able to transduce proliferative signals on CD30+ target cells, including cultured H-RS cells. Our data suggest that eosinophils may not merely represent innocent bystanders, but rather act as important elements in the pathology of HD by contributing to the deregulated network of CD30/CD30L-mediated interactive signals between H-RS cells and surrounding reactive cells.
APA, Harvard, Vancouver, ISO, and other styles
16

Saraiva, Margarida, Philip Smith, Padraic G. Fallon, and Antonio Alcami. "Inhibition of Type 1 Cytokine–mediated Inflammation by a Soluble CD30 Homologue Encoded by Ectromelia (Mousepox) Virus." Journal of Experimental Medicine 196, no. 6 (September 16, 2002): 829–39. http://dx.doi.org/10.1084/jem.20020319.

Full text
Abstract:
CD30 is up-regulated in several human diseases and viral infections but its role in immune regulation is poorly understood. Here, we report the expression of a functional soluble CD30 homologue, viral CD30 (vCD30), encoded by ectromelia (mousepox) virus, a poxvirus that causes a severe disease related to human smallpox. We show that vCD30 is a 12-kD secreted protein that not only binds CD30L with high affinity and prevents its interaction with CD30, but it also induces reverse signaling in cells expressing CD30L. vCD30 blocked the generation of interferon γ–producing cells in vitro and was a potent inhibitor of T helper cell (Th)1- but not Th2-mediated inflammation in vivo. The finding of a CD30 homologue encoded by ectromelia virus suggests a role for CD30 in antiviral defense. Characterization of the immunological properties of vCD30 has uncovered a role of CD30–CD30L interactions in the generation of inflammatory responses.
APA, Harvard, Vancouver, ISO, and other styles
17

Gattei, Valter, Massimo Degan, Annunziata Gloghini, Angela De Iuliis, Salvatore Improta, Francesca Maria Rossi, Donatella Aldinucci, et al. "CD30 Ligand Is Frequently Expressed in Human Hematopoietic Malignancies of Myeloid and Lymphoid Origin." Blood 89, no. 6 (March 15, 1997): 2048–59. http://dx.doi.org/10.1182/blood.v89.6.2048.

Full text
Abstract:
Abstract CD30 ligand (CD30L) is a type-II membrane glycoprotein capable of transducing signals leading to either cell death or proliferation through its specific counterstructure CD30. Although several lines of evidence indicate that CD30L plays a key role as a paracrine- or autocrine-acting surface molecule in the deregulated cytokine cascade of Hodgkin's disease, little is known regarding its distribution and biologic significance in other human hematopoietic malignancies. By analyzing tumor cells from 181 patients with RNA studies and immunostaining by the anti-CD30L monoclonal antibody M80, we were able to show that human hematopoietic malignancies of different lineage and maturation stage display a frequent and broad expression of the ligand. CD30L mRNA and surface protein were detected in 60% of acute myeloid leukemias (AMLs), 54% of B-lineage acute lymphoblastic leukemias (ALLs), and in a consistent fraction (68%) of B-cell lymphoproliferative disorders. In this latter group, hairy cell leukemia and high-grade B-cell non-Hodgkin's lymphoma (B-NHL) expressed a higher surface density of CD30L as compared with B-cell chronic lymphocytic leukemia and low-grade B-NHL. Purified plasmacells from a fraction of multiple myeloma patients also displayed CD30L mRNA and protein. A more restricted expression of CD30L was found in T-cell tumors that was mainly confined to neoplasms with an activated peripheral T-cell phenotype, such as T-cell prolymphocytic leukemia, peripheral T-NHL, and adult T-cell leukemia/lymphoma. In contrast, none of the T-lineage ALLs analyzed expressed the ligand. In AML, a high cellular density of CD30L was detected in French-American-British M3, M4, and M5 phenotypes, which are directly associated with the presence on tumor cells of certain surface structures, including the p55 interleukin-2 receptor α-chain, the αM (CD11b) chain of β2 integrins, and the intercellular adhesion molecule-1 (CD54). Analysis of normal hematopoietic cells evidenced that, in addition to circulating and tonsil B cells, a fraction of bone marrow myeloid precursors, erythroblasts, and subsets of megakaryocytes also express CD30L. Finally, we have shown that native CD30L expressed on primary leukemic cells is functionally active by triggering both mitogenic and antiproliferative signals on CD30+ target cells. As opposed to CD30L, only 10 of 181 primary tumors expressed CD30 mRNA or protein, rendering therefore unlikely a CD30-CD30L autocrine loop in human hematopoietic neoplasms. Taken together, our data indicate that CD30L is widely expressed from early to late stages of human hematopoiesis and suggest a regulatory role for this molecule in the interactions of normal and malignant hematopoietic cells with CD30+ immune effectors and/or microenvironmental accessory cells.
APA, Harvard, Vancouver, ISO, and other styles
18

Jiao, Yingying, Xiaoyuan Zeng, Zongpeng Li, Beng H. Chong, Yujiao Zhang, and Jieyu Ye. "Role of Thrombopoietin in Stimulating Megakaryopoiesis of ITP Patients Via Rescuing Vascular Niche." Blood 134, Supplement_1 (November 13, 2019): 1063. http://dx.doi.org/10.1182/blood-2019-125679.

Full text
Abstract:
Background: Previous studies have shown that the vascular niche was damaged in patients with immune thrombocytopenia (ITP). It is mainly manifested that the migration and tube-formation ability of bone marrow endothelial progenitor cells (BM-EPCs) were significantly reduced. Endothelial cells as an important part of the vascular niche play an important role in regulation of megakaryocytes and platelet hematopoiesis. Our previous studies suggested that thrombopoietin (TPO) has a protective effect on human endothelial cells. This study aim to investigate whether TPO has a protective effect in EPCs of ITP patients and to explore its mechanism. Methods: BM-EPCs were isolated from ITP patients, and divided into two groups: TPO-treated group and TPO-free (control group). CCK8 was used to explore whether TPO has proliferative effect on BM-EPCs. BM-EPCs was identified by flow cytometry with co-expression of mouse anti-humanCD133, mouse anti-human CD34 and mouse anti-human vascular endothelial growth factor receptor (CD309) monoclonal antibodies. Moreover, the number of EPCs was evaluated with DiL-Ac-LDL uptake and FITC-UEA -I binding assay. Furthermore, the function of BM-EPCs was studied by using tube-formation and migration experiments. Results: The ITP patient-derived EPCs in TPO-treated group exhibited longer fusiform and elliptical shape after 5 days and cobblestone-like morphology after 7-10 days. The result of CCK-8 showed TPO could promote the proliferation of EPCs and the optional drug concentration is 100 ng/ml. Much higher expression of CD34/CD133/CD309 in TPO-treated group was detected by flow cytometry. The number of cells with immunofluorescence double staining in TPO group was significantly higher than of control group (n=4, p<0.05). The tube formation (n=5, p<0.05) and migration capacity (n=5, p<0.05) were significantly enhanced in TPO-treated group. Conclusion: Our studies indicates that TPO may exert megakaryopoiesis effect in ITP patients via protection of BM-EPCs. This may provide a supplemental explanation of how recombinant TPO and TPO receptor antagonist acts on ITP treatment. Keywords: thrombopoietin; vascular niche; endothelial progenitor cell, megakaryopoiesis; endothelial cell Disclosures No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
19

Luo, Liang, Yangli Liu, Dubo Chen, Fengjia Chen, Hai Bing Lan, and Canmao Xie. "CD30 Is Highly Expressed in Chronic Obstructive Pulmonary Disease and Induces the Pulmonary Vascular Remodeling." BioMed Research International 2018 (June 10, 2018): 1–13. http://dx.doi.org/10.1155/2018/3261436.

Full text
Abstract:
Chronic obstructive pulmonary disease (COPD) is one of the common and underdiagnosed diseases with the highest morbidity and mortality in the world. The development of COPD can lead to pulmonary vascular remodeling and pulmonary hypertension, further causing the occurrence of pulmonary heart disease. Therefore, attenuation of pulmonary vascular remodeling and pulmonary hypertension caused by COPD can significantly delay cardiovascular complications. In the study, we firstly found that the expression of CD30 and CD30L was increased in COPD. Importantly, the serum CD30L levels were significantly higher in patients with stable COPD relative to those with acute exacerbation of COPD (AECOPD). This suggested that CD30 might be related to the development of COPD. In addition, we found that the expression of CD30 in the COPD rat model was significantly increased compared with control group. And treatment with the anti-CD30 antibody reduced the serum concentration and tissue expression of CD30 in rat. Importantly, anti-CD30 antibody alleviated pulmonary vascular remodeling in COPD model rats. This suggested that CD30 played an important role in the course of COPD. Finally, we found that, in the HPASMC and HPAEC cell lines, CD30 can affect the cell viability and cell migration and inhibited hypoxia-induced cell apoptosis in a concentration-dependent manner. We also found CD30 induced extracellular matrix formation through decreasing the expression of MMP-2, thus promoting the pulmonary vascular remodeling. The study indicated that CD30 and CD30L were involved in pulmonary vascular remodeling and inflammatory response in COPD. Altogether, CD30 might be a marker for the early diagnosis and progression of COPD.
APA, Harvard, Vancouver, ISO, and other styles
20

Dehghani, Leila, Hamid Ganji, Nahid Eskandari, Reza Soleimani, Maedeh Talebi, FazelSahraneshin Samani, Rokhsareh Meamar, and Masoud Etemadifar. "Evaluation of the circulating CD34+, CD309+, and endothelial progenitor cells in patients with first attack of optic neuritis." Advanced Biomedical Research 4, no. 1 (2015): 151. http://dx.doi.org/10.4103/2277-9175.161578.

Full text
APA, Harvard, Vancouver, ISO, and other styles
21

Першина, О. В., А. В. Пахомова, Н. Н. Ермакова, О. Ю. Рыбалкина, В. А. Крупин, Э. С. Пан, О. Е. Ваизова, et al. "Circulating stem and progenitor cells as markers of inflammation, endothelial regeneration, and prediction of vascular complications in metabolic syndrome and type 1 and 2 diabetes mellitus." Nauchno-prakticheskii zhurnal «Patogenez», no. 1() (March 20, 2018): 58–67. http://dx.doi.org/10.25557/2310-0435.2018.01.58-67.

Full text
Abstract:
Цель исследования состояла в выявлении информативных клеточных маркеров сосудистых осложнений, регенерации микрососудистой сети и воспаления в венозной крови здоровых волонтеров, больных с метаболическим синдромом, сахарным диабетом 1 и 2 типа. Методы. Обследованы больные с метаболическим синдромом (МС), диабетом 2 типа без осложнений, диабетом 1 типа средней степени тяжести и здоровые волонтеры. Диагноз пациентов подтвержден общеклиническими, биохимическими, коагулометрическими и иммуноферментными методами исследования, для оценки экспрессии антигенов использовался многопараметрический цитометрический анализ. Результаты. При анализе экспрессии маркеров показано изменение числа эндотелиальных клеток, мезенхимальных стволовых клеток (МСК) и гемопоэтических стволовых клеток (ГСК) в крови в зависимости от патологии. Эндотелиальные клетки миелоидного (CD45CD14CD34CD309CD144CD31) и немиелоидного (CD45CD14CD34CD309CD144CD31) происхождения, CD309-эндотелиальные клетки и МСК (CD44CD73CD90CD105) предлагаются в качестве маркеров повреждения эндотелия при диабетической симптоматике. При этом ГСК (CD45CD34) могут выступать ценным диагностическим и прогностическим маркером воспаления. Заключение. Для подтверждения сосудистых повреждений и прогноза развития осложнений при диабете 1 и 2 типа в венозной крови пациентов целесообразно оценивать эндотелиальные прогениторные клетки (ЭПК) не костномозговой локализации (CD31CD309CD144) и костномозговой локализации (CD34CD309), и ЭПК c высоким регенеративным потенциалом (CD45CD34CD31CD144). Циркулирующие ЭПК, формирующие колонии in vitro (CD45CD34CD31), рекомендуется использовать в качестве дифференциального маркера состояния регенерации эндотелия при диабете 2 типа. The aim of this study was to identify mesenchymal stem cells (MSC), hematopoietic stem cells (HSC), mature endothelial cells, and endothelial progenitor cells (EPC) in the blood of healthy volunteers, patients with metabolic syndrome, and type 1 and 2 diabetes mellitus as new, informative cellular markers of vascular complications, endothelial regeneration, and inflammation. Methods. The diagnosis was confirmed by general clinical, biochemical, coagulometeric and ELISA studies; multi-parameter cytometric assay was used for evaluation of antigen expression. Results. Changes in the count of MSC, HSC, mature endothelial cells, and endothelial progenitor cells in blood of patients with metabolic syndrome and type 1 and 2 diabetes depended on the type of pathology. We propose using endothelial cells of myeloid (CD45CD14CD34CD309CD144CD31) and non-myeloid origin (CD45CD14CD34CD309CD144CD31), CD309-endothelial cells, and MSCs with the CD44CD73CD90CD105 phenotype as nonspecific markers of endothelial damage in presence of diabetic symptoms. Furthermore, HSCs (CD45CD34) can be used as a valuable diagnostic and prognostic marker of inflammation. Conclusions. It is relevant to evaluate EPCs of non-bone marrow localization (CD31CD309CD144) and bone marrow localization (CD34CD309) and EPCs with a high regenerative potential (CD45CD34CD31CD144) in the blood of patients with type 1 and 2 diabetes to confirm the presence of vascular damage and predict development of complications. Circulating, in vitro colony-forming EPCs (CD45CD34CD31) are recommended as a differential marker for inhibition of endothelial regeneration in type 2 diabetes.
APA, Harvard, Vancouver, ISO, and other styles
22

Fernandes, Eloise Helena, Alberto José Prioli, Carlos Alberto Scapim, Ivan Schuster, Elisa Serra Negra Vieira, Antônio Teixeira do Amaral Júnior, and Lia Mara Moterle. "Embriogênese somática a partir de embriões imaturos em genótipos de milho." Ciência Rural 38, no. 9 (December 2008): 2604–7. http://dx.doi.org/10.1590/s0103-84782008000900031.

Full text
Abstract:
A primeira etapa de um programa de transformação genética de plantas é o estabelecimento de um protocolo de regeneração de plantas, a partir de cultura de tecidos. A regeneração de plantas pode ser conseguida por meio de organogênese ou embriogênese. A embriogênese somática é dependente do genótipo das plantas utilizadas e a identificação de genótipos mais responsivos a este método de regeneração resulta em maior eficiência da técnica. O trabalho teve como objetivo identificar genótipos de milho com maior capacidade de produção de embriões somáticos e regeneração de plantas. Foram investigados 11 genótipos (linhagens e híbridos). As culturas foram obtidas a partir de embriões imaturos, inoculados em meio N6, suplementado com 690mg L-1 de prolina e 10mM de 2,4-D, com subcultivos quinzenais. Nos genótipos LD82025, CD308 e CML314, foram observados calos do tipo II, friáveis e embriogênicos. Esses genótipos foram submetidos ao processo de regeneração, destacando-se a linhagem LD82025. Os genótipos CD307, CD304, OC-705, 105-B e o GU04328 não apresentaram indução de calos embriogênicos. Os resultados indicam que a linhagem LD82025 é a mais promissora para utilização em um programa de transformação genética de plantas.
APA, Harvard, Vancouver, ISO, and other styles
23

Ansheles, A. A., A. V. Rvacheva, and I. V. Sergienko. "Effect of Atorvastatin Therapy on the Level of CD34+CD133+CD309+ Endothelial Progenitor Cells in Patients with Coronary Heart Disease." Bulletin of Experimental Biology and Medicine 163, no. 1 (May 2017): 133–36. http://dx.doi.org/10.1007/s10517-017-3753-7.

Full text
APA, Harvard, Vancouver, ISO, and other styles
24

Zbucka-Kretowska, Monika, Andrzej Eljaszewicz, Danuta Lipinska, Kamil Grubczak, Malgorzata Rusak, Grzegorz Mrugacz, Milena Dabrowska, Mariusz Z. Ratajczak, and Marcin Moniuszko. "Effective Mobilization of Very Small Embryonic-Like Stem Cells and Hematopoietic Stem/Progenitor Cells but Not Endothelial Progenitor Cells by Follicle-Stimulating Hormone Therapy." Stem Cells International 2016 (2016): 1–8. http://dx.doi.org/10.1155/2016/8530207.

Full text
Abstract:
Recently, murine hematopoietic progenitor stem cells (HSCs) and very small embryonic-like stem cells (VSELs) were demonstrated to express receptors for sex hormones including follicle-stimulating hormone (FSH). This raised the question of whether FSH therapy at clinically applied doses can mobilize stem/progenitor cells in humans. Here we assessed frequencies of VSELs (referred to as Lin−CD235a−CD45−CD133+cells), HSPCs (referred to as Lin−CD235a−CD45+CD133+cells), and endothelial progenitor cells (EPCs, identified as CD34+CD144+, CD34+CD133+, and CD34+CD309+CD133+cells) in fifteen female patients subjected to the FSH therapy. We demonstrated that FSH therapy resulted in statistically significant enhancement in peripheral blood (PB) number of both VSELs and HSPCs. In contrast, the pattern of responses of EPCs delineated by different cell phenotypes was not uniform and we did not observe any significant changes in EPC numbers following hormone therapy. Our data indicate that FSH therapy mobilizes VSELs and HSPCs into peripheral blood that on one hand supports their developmental origin from germ lineage, and on the other hand FSH can become a promising candidate tool for mobilizing HSCs and stem cells with VSEL phenotype in clinical settings.
APA, Harvard, Vancouver, ISO, and other styles
25

Zambello, Renato, Livio Trentin, Monica Facco, Marta Siviero, Silvia Galvan, Francesco Piazza, Alessandra Perin, Carlo Agostini, and Gianpietro Semenzato. "Analysis of TNF-receptor and ligand superfamily molecules in patients with lymphoproliferative disease of granular lymphocytes." Blood 96, no. 2 (July 15, 2000): 647–54. http://dx.doi.org/10.1182/blood.v96.2.647.014k18_647_654.

Full text
Abstract:
In 21 patients with lymphoproliferative disease of granular lymphocytes (LDGL), we investigated the expression and the function of molecules belonging to TNF-receptor and TNF-ligand superfamilies (CD30/CD30L; CD40/CD40L; CD27/CD70; Fas [CD95]/FasL[CD95L]). Fourteen patients were characterized by a proliferation of granular lymphocytes (GLs) expressing the CD3+CD16+phenotype, whereas 7 cases showed the CD3−CD16+ CD56 ± phenotype. Our data show that both CD3+ and CD3-GLs are preferentially equipped with CD30, CD40, CD40L, CD70, and CD95 antigens; this pattern is usually associated with the lack of CD27 and CD30L antigens expression. CD95L was demonstrated in the cytoplasm in 14 of 21 cases by flow cytometry, but a definite signal was demonstrated in all cases studied using polymerase chain reaction analysis. On functional grounds, a stimulatory activity on rIL-2 mediated redirected-cytotoxicity against Fcγ+ P815 targets was demonstrated with anti-CD30, CD40, CD40L, CD70, CD95, and CD95L mAbs, although resting cells were unable to exhibit significant redirected-cell lysis. The addition of anti-CD30, CD30L, CD40, CD40L, CD95, and CD95L mAbs did not show any significant effect on cell proliferation at resting conditions or after rIL-2 stimulation, whereas anti-CD70 mAb mediated cell proliferation in 6 of 10 cases tested. This figure was not related to an increase in apoptotic cells, as investigated by Annexin-V expression. Our data indicate that both CD3+ and CD3− GLs are equipped with different costimulatory antigens, supporting the concept that these cells are in vivo activated and suggesting that these molecules might play a role in the cytotoxic mechanisms of GLs.
APA, Harvard, Vancouver, ISO, and other styles
26

Zambello, Renato, Livio Trentin, Monica Facco, Marta Siviero, Silvia Galvan, Francesco Piazza, Alessandra Perin, Carlo Agostini, and Gianpietro Semenzato. "Analysis of TNF-receptor and ligand superfamily molecules in patients with lymphoproliferative disease of granular lymphocytes." Blood 96, no. 2 (July 15, 2000): 647–54. http://dx.doi.org/10.1182/blood.v96.2.647.

Full text
Abstract:
Abstract In 21 patients with lymphoproliferative disease of granular lymphocytes (LDGL), we investigated the expression and the function of molecules belonging to TNF-receptor and TNF-ligand superfamilies (CD30/CD30L; CD40/CD40L; CD27/CD70; Fas [CD95]/FasL[CD95L]). Fourteen patients were characterized by a proliferation of granular lymphocytes (GLs) expressing the CD3+CD16+phenotype, whereas 7 cases showed the CD3−CD16+ CD56 ± phenotype. Our data show that both CD3+ and CD3-GLs are preferentially equipped with CD30, CD40, CD40L, CD70, and CD95 antigens; this pattern is usually associated with the lack of CD27 and CD30L antigens expression. CD95L was demonstrated in the cytoplasm in 14 of 21 cases by flow cytometry, but a definite signal was demonstrated in all cases studied using polymerase chain reaction analysis. On functional grounds, a stimulatory activity on rIL-2 mediated redirected-cytotoxicity against Fcγ+ P815 targets was demonstrated with anti-CD30, CD40, CD40L, CD70, CD95, and CD95L mAbs, although resting cells were unable to exhibit significant redirected-cell lysis. The addition of anti-CD30, CD30L, CD40, CD40L, CD95, and CD95L mAbs did not show any significant effect on cell proliferation at resting conditions or after rIL-2 stimulation, whereas anti-CD70 mAb mediated cell proliferation in 6 of 10 cases tested. This figure was not related to an increase in apoptotic cells, as investigated by Annexin-V expression. Our data indicate that both CD3+ and CD3− GLs are equipped with different costimulatory antigens, supporting the concept that these cells are in vivo activated and suggesting that these molecules might play a role in the cytotoxic mechanisms of GLs.
APA, Harvard, Vancouver, ISO, and other styles
27

Donahue, Robert E., Ping Jin, Naoya Uchida, Aylin C. Bonifacino, Yu Tian, Johanna I. Klinman, Joseph J. Mattapallil, et al. "CD34+CXCR4(CD184)+ Cells Differentiate Into Myeloid Dendritic Cell Progenitors." Blood 122, no. 21 (November 15, 2013): 4835. http://dx.doi.org/10.1182/blood.v122.21.4835.4835.

Full text
Abstract:
Abstract Dendritic cells (DCs) play a central role in innate immunity and adaptive immunity. DCs are antigen presenting cells capable of inducing primary T cell responses or facilitating self-tolerance. Myeloid DCs (mDC) express CD11c. They are further divided into Type 1 myeloid DCs (mDC1) which are CD1c+CD141+ and Type 2 mDCs (mDC2) which are CD1c-CD141+, and plasmacytoid DC which are CD11c- and express CD303. Plasmacytoid DCs are the main source of type 1 interferon upon infection. CD34+ cells are a heterogeneous population of cells which contain both hematopoietic progenitors and stem cells. The microarray signature of CD34+CXCR4 (CD184)+ cells in both human and non-human primates suggest that this cell population plays a role in innate immunity. The signaling pathway for the Triggering Receptor Expressed on Myeloid Cells 1 (TREM1) is the most up-regulated pathway in both human and non-human primate sorted CD34+CD184+ cells. The TREM family is involved in the amplification and regulation of inflammatory responses. Upon sorting both human and rhesus CD34+ cells into CD34+CD184+ and CD34+CD184-, we observe that CD34+CD184+ cells are both adherent and non-adherent in nature (Figure 1) and while maintained in Flt3-L, IL-3, and SCF form progeny which appear dendritic in nature (Figure 2). In addition, we have found the CD34+CD184+ subpopulation to be resistant to lentiviral vector transduction. Upon culturing in defined, serum free media supplemented with cytokines, the progeny of the CD34+CD184+ population using both flow cytometry and confocal microscopy are CD11c+ and contain both CD1c+ and CD1c- subpopulations (Figure 3) with a predominately CD80(B7-1)+, CD123(IL-3R)+, CD197 (CCR7)+ , and HLA-Dr+ immunophenotype, having variability in CD86(B7-2) +/-, CD141(BDCA-3/Thrombomodulin)+/- , and CD144 (VE-cadherin)+/- expression, and being negative for CD303(BDCA-2)and CD309(VEGFR). Of special interest is that the CD34+CD184+ progeny contain both CD1c-CD16+ and CD1c+CD16- subpopulations of mDC. CD1c-CD16+ mDC have been shown to have strong pro-inflammatory activity, whereas CD1c+CD16- mDC are mainly inducers of chemotaxis. The progeny of the CD34+CD184+ cells can be stimulated by LPS and IFNγ to produce IL-12 based on an enzyme-linked immunosorbent assay. These results demonstrate the importance of the CD34+CXCR4+ progenitor in mDC development and allow one to speculate on how this mDC progenitor might prove of therapeutic benefit in vaccine development and cancer therapy.Figure 1Human CD34+CD184+ Sorted PopulationFigure 1. Human CD34+CD184+ Sorted PopulationFigure 2Cultured Progeny CD34+CD184+Figure 2. Cultured Progeny CD34+CD184+Figure 3Non-Human Primate CD34+CD184+ Confocal MicroscopyFigure 3. Non-Human Primate CD34+CD184+ Confocal Microscopy Disclosures: No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
28

Thakar, Nilay Y., Dmitry A. Ovchinnikov, Marcus L. Hastie, Bostjan Kobe, Jeffrey J. Gorman, and Ernst J. Wolvetang. "TRAF2 recruitment via T61 in CD30 drives NFκB activation and enhances hESC survival and proliferation." Molecular Biology of the Cell 26, no. 5 (March 2015): 993–1006. http://dx.doi.org/10.1091/mbc.e14-08-1290.

Full text
Abstract:
CD30 activates NFκB signaling in human embryonic stem cells. A single threonine residue in the CD30v protein is critical for this and recruitment of TRAF2. The data reveal the importance of this interaction for hESC survival and proliferation.
APA, Harvard, Vancouver, ISO, and other styles
29

Pulito-Cueto, Verónica, Sara Remuzgo-Martínez, Fernanda Genre, Víctor M. Mora-Cuesta, David Iturbe-Fernández, Sonia Fernández-Rozas, Belén Atienza-Mateo, et al. "Endothelial Progenitor Cells as a Potential Biomarker in Interstitial Lung Disease Associated with Rheumatoid Arthritis." Journal of Clinical Medicine 9, no. 12 (December 18, 2020): 4098. http://dx.doi.org/10.3390/jcm9124098.

Full text
Abstract:
Interstitial lung disease (ILD) increases morbidity and mortality in patients with rheumatoid arthritis (RA). Although the pathogenesis of ILD associated with RA (RA-ILD+) remains poorly defined, vascular tissue is crucial in lung physiology. In this context, endothelial progenitor cells (EPC) are involved in endothelial tissue repair. However, little is known about their implication in RA-ILD+. Accordingly, we aimed to investigate the potential role of EPC related to endothelial damage in RA-ILD+. EPC quantification in peripheral blood from 80 individuals (20 RA-ILD+ patients, 25 RA-ILD− patients, 21 idiopathic pulmonary fibrosis (IPF) patients, and 14 healthy controls) was performed by flow cytometry. EPC were considered as CD34+, CD45low, CD309+ and CD133+. A significant increase in EPC frequency in RA-ILD+ patients, as well as in RA-ILD− and IPF patients, was found when compared with controls (p < 0.001, p = 0.02 and p < 0.001, respectively). RA-ILD+ patients exhibited a higher EPC frequency than the RA-ILD− ones (p = 0.003), but lower than IPF patients (p < 0.001). Our results suggest that EPC increase may represent a reparative compensatory mechanism in patients with RA-ILD+. The degree of EPC frequency may help to identify the presence of ILD in RA patients and to discriminate RA-ILD+ from IPF.
APA, Harvard, Vancouver, ISO, and other styles
30

Alvarez, Diego F., Lan Huang, Judy A. King, M. Khair ElZarrad, Mervin C. Yoder, and Troy Stevens. "Lung microvascular endothelium is enriched with progenitor cells that exhibit vasculogenic capacity." American Journal of Physiology-Lung Cellular and Molecular Physiology 294, no. 3 (March 2008): L419—L430. http://dx.doi.org/10.1152/ajplung.00314.2007.

Full text
Abstract:
Endothelial progenitor cells (EPCs) have been isolated postnatally from bone marrow, blood, and both the intima and adventitia of conduit vessels. However, it is unknown whether EPCs can be isolated from the lung microcirculation. Thus we sought to determine whether the microvasculature possesses EPCs capable of de novo vasculogenesis. Rat pulmonary artery (PAEC) and microvascular (PMVEC) endothelial cells were isolated and selected by using a single-cell clonogenic assay. Whereas the majority of PAECs (∼60%) were fully differentiated, the majority of PMVECs (∼75%) divided, with ∼50% of the single cells giving rise to large colonies (>2,000 cells/colony). These highly proliferative cells exhibited the capacity to reconstitute the entire proliferative hierarchy of PMVECs, unveiling the existence of resident microvascular endothelial progenitor cells (RMEPCs). RMEPCs expressed endothelial cell markers (CD31, CD144, endothelial nitric oxide synthase, and von Willenbrand factor) and progenitor cell antigens (CD34 and CD309) but did not express the leukocyte marker CD45. Consistent with their origin, RMEPCs interacted with Griffonia simplicifolia and displayed restrictive barrier properties. In vitro and in vivo Matrigel assays revealed that RMEPCs possess vasculogenic capacity, forming ultrastructurally normal de novo vessels. Thus the pulmonary microcirculation is enriched with EPCs that display vasculogenic competence while maintaining functional endothelial microvascular specificity.
APA, Harvard, Vancouver, ISO, and other styles
31

Laupheimer, Michael, Anna Skorska, Jana Große, Gudrun Tiedemann, Gustav Steinhoff, Robert David, and Cornelia A. Lux. "Selective Migration of Subpopulations of Bone Marrow Cells along an SDF-1α and ATP Gradient." Bone Marrow Research 2014 (December 31, 2014): 1–10. http://dx.doi.org/10.1155/2014/182645.

Full text
Abstract:
Both stem cell chemokine stromal cell-derived factor-1α (SDF-1α) and extracellular nucleotides such as adenosine triphosphate (ATP) are increased in ischemic myocardium. Since ATP has been reported to influence cell migration, we analysed the migratory response of bone marrow cells towards a combination of SDF-1 and ATP. Total nucleated cells (BM-TNCs) were isolated from bone marrow of cardiac surgery patients. Migration assays were performed in vitro. Subsequently, migrated cells were subjected to multicolor flow cytometric analysis of CD133, CD34, CD117, CD184, CD309, and CD14 expression. BM-TNCs migrated significantly towards a combination of SDF-1 and ATP. The proportions of CD34+ cells as well as subpopulations coexpressing multiple stem cell markers were selectively enhanced after migration towards SDF-1 or SDF-1 + ATP. After spontaneous migration, significantly fewer stem cells and CD184+ cells were detected. Direct incubation with SDF-1 led to a reduction of CD184+ but not stem cell marker-positive cells, while incubation with ATP significantly increased CD14+ percentage. In summary, we found that while a combination of SDF-1 and ATP elicited strong migration of BM-TNCs in vitro, only SDF-1 was responsible for selective attraction of hematopoietic stem cells. Meanwhile, spontaneous migration of stem cells was lower compared to BM-TNCs or monocytes.
APA, Harvard, Vancouver, ISO, and other styles
32

Foris, Vasile, Gabor Kovacs, Leigh M. Marsh, Zoltán Bálint, Martin Tötsch, Alexander Avian, Philipp Douschan, et al. "CD133+ cells in pulmonary arterial hypertension." European Respiratory Journal 48, no. 2 (April 21, 2016): 459–69. http://dx.doi.org/10.1183/13993003.01523-2015.

Full text
Abstract:
Circulating mononuclear cells may play an important role for the vascular remodelling in pulmonary arterial hypertension (PAH), but studies addressing multiple progenitor populations are rare and inconsistent.We used a comprehensive fluorescence-activated cell sorting analysis of circulating mononuclear cells in 20 PAH patients and 20 age- and sex-matched controls, and additionally analysed CD133+ cells in the lung tissue of five PAH transplant recipients and five healthy controls (donor lungs).PAH patients were characterised by increased numbers of circulating CD133+ cells and lymphopenia as compared with control. In PAH, CD133+ subpopulations positive for CD117 or CD45 were significantly increased, whereas CD133+CD309+, CD133+CXCR2+ and CD133+CD31+ cells were decreased. In CD133+ cells, SOX2, Nanog, Ki67 and CXCR4 were not detected, but Oct3/4 mRNA was present in both PAH and controls. In the lung tissue, CD133+ cells included three main populations: type 2 pneumocytes, monocytes and undifferentiated cells without significant differences between PAH and controls.In conclusion, circulating CD133+ progenitor cells are elevated in PAH and consist of phenotypically different subpopulations that may be up- or downregulated. This may explain the inconsistent results in the literature. CD133+ type 2 pneumocytes in the lung tissue are not associated with circulating CD133+ mononuclear cells.
APA, Harvard, Vancouver, ISO, and other styles
33

Sanda, Naomi, Nobuaki Suzuki, Mayuko Kishimoto, Natsumi Kameyama, Yoko Kajiura, Hiroyuki Matsumoto, Shigeo Nakamura, and Tadashi Matsushita. "Establishment of Blood Outgrowth Endothelial Cells from a Type 3 Von Willebrand Disease (VWD) Patient with a Neutralizing Anti-VWF Inhibitors." Blood 124, no. 21 (December 6, 2014): 1508. http://dx.doi.org/10.1182/blood.v124.21.1508.1508.

Full text
Abstract:
Abstract Background: Type 3 von Willebrand disease (VWD) is characterized by a complete loss type of von Willebrand Factor (VWF) with the rarest disease frequency and hemorrhagic symptom is the most severe among other VWD types. The development of alloantibodies directed against VWF occurs in approximately 10% of patients with type 3 VWD. In this study, we found the VWF gene alterations and established blood outgrowth endothelial cells (BOECs) a from a Japanese type 3 VWD patient with an anti-VWF inhibitors. Case: A 5-year-old woman suffered from epistaxis, purpura and easy bruising and has been diagnosed as VWD. Her plasma level of FVIII:C was 1.8% and VWF:Ag and VWF:RCo was <5% and <10%, respectively. Her plasma lacked VWF multimers, indicating that she had type 3 VWD. To treat her frequent muco-cutaneous bleeding, purified plasma-derived FVIII/VWF concentrates were administrated followed by inhibitor development. After about 20ED of FVIII/VWF concentrates, an anaphylactic symptom comprised of cough, dyspnea, and wheezing developed at the time of dosing. An high titer inhibitory antibody against VWF was confirmed by a Bethesda assay based on a assay of VWF:RCo. Her younger sister also had type 3 VWD. Methods: Patients samples were collected after the written informed consent has bee obtained. We performed MLPA for genetic deletion or insertion. Then we amplified the all exons including the exon/intron boundaries of the VWF gene by PCR using allele-specific primers, and analyzed DNA sequences of the patient. Peripheral blood mononuclear cells were obtained from the patient and BOECs were then established. Briefly, buffy coat mononuclear cells were isolated and seeded onto a 6-well tissue culture plate precoated with type 1 rat tail collagen at 37°C, 5% CO2, in a humidified incubator. Medium was changed daily for 7 days and then every other day until the first passage. The endothelial identity of the BOECs was confirmed by flow cytometric and immunofluorescence analyses using endothelial markers with antibodies to CD31, VE-Cadherin (CD144) and VEGFR-2 (CD309). Results: The large deletion and insertion of VWF gene were not found. Direct sequencing showed the propositus and her sister were compound heterozygous for an E2341X (c.7021G>T) mutation in exon41 and a Y2631X (c.7892-7893insA) mutation in exon48. These were therefore two novel nonsense mutations and normal VWF polypeptides could not be translated. Flow cytometric analysis indicated that established BOECs expressed cell surface CD31 and CD309, whereas CD34 was not detected. By immunofluorescence analysis of fixed BOECs, VWF signal was remarkably reduced from the patient, in spite of normal VE-Cadherin expression. VWF specific ELISA was performed for both cell supernatant and lysates of patient’s BOECs but immunodetectable VWF was not secreted. Discussion: We identified two novel nonsense mutations causing type3 VWD. BOECs established from the patient reproduced the phenotype of the disease, suggesting that BOEC analysis is useful for studying the molecular pathogenesis of von Willebrand disease. Disclosures No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
34

Foks, Amanda C., Ilze Bot, Vanessa Frodermann, Saskia C. A. de Jager, Mariette ter Borg, Peter J. van Santbrink, Hideo Yagita, Johan Kuiper, and Gijs H. M. van Puijvelde. "Interference of the CD30–CD30L Pathway Reduces Atherosclerosis Development." Arteriosclerosis, Thrombosis, and Vascular Biology 32, no. 12 (December 2012): 2862–68. http://dx.doi.org/10.1161/atvbaha.112.300509.

Full text
APA, Harvard, Vancouver, ISO, and other styles
35

De Lorenzo, Andrea, Annie S. B. Moreira, Fabiana B. Muccillo, Marcelo Assad, and Eduardo V. Tibirica. "Microvascular Function and Endothelial Progenitor Cells in Patients with Severe Hypercholesterolemia and the Familial Hypercholesterolemia Phenotype." Cardiology 137, no. 4 (2017): 231–36. http://dx.doi.org/10.1159/000470829.

Full text
Abstract:
Objective: To evaluate endothelial progenitor cells (EPCs) and systemic microvascular function in patients with severe hypercholesterolemia, comparing patients with the definite familial hypercholesterolemia (FH) phenotype (DFH) or probable/possible FH phenotype (PFH). There is a large spectrum of atherosclerotic disease between these two clinical phenotypes of FH, and to acquire further knowledge of the pathophysiology of vascular disease in both is desirable. Methods: Subjects with severe hypercholesterolemia, defined as low-density lipoprotein cholesterol (LDL-C) >190 mg/dL, were classified as DFH or PFH and underwent measurement of the number of EPCs by flow cytometry and evaluation of cutaneous microvascular reactivity using a laser speckle contrast-imaging system with iontophoresis of acethylcholine (ACh) or sodium nitroprusside. EPCs were defined as CD45- or CD45low, CD34+CD133+CD309+ cells. Categorical variables were compared using Fisher test and continuous variables with Student t test or Mann-Whitney test, and a value of p < 0.05 was considered statistically significant. Results: Patients with DFH had higher LDL-C than those with PFH. There was no difference in the median number of EPCs between patients with DFH or PFH, but there was a significant reduction of endothelial-dependent, ACh-induced vasodilatation in the former. Conclusion: Patients with DFH have impaired microvascular endothelial-dependent vasodilatation compared to those with PFH, indicating more severe vascular disease in the former.
APA, Harvard, Vancouver, ISO, and other styles
36

Gubar, O. S., A. I. Rodnichenko, R. G. Vasylie, A. V. Zlatska, and D. O. Zubov. "POSTNATAL EXTRA-EMBRYONIC TISSUES AS A SOURCE OF MULTIPLE CELL TYPES FOR REGENERATIVE MEDICINE APPLICATIONS." Experimental Oncology 39, no. 3 (September 22, 2017): 186–90. http://dx.doi.org/10.31768/2312-8852.2017.39(3):186-190.

Full text
Abstract:
Aim: We aimed to isolate and characterize the cell types which could be obtained from postnatal extra-embryonic tissues. Materials and Methods: Fresh tissues (no more than 12 h after delivery) were used for enzymatic or explants methods of cell isolation. Obtained cultures were further maintained at 5% oxygen. At P3 cell phenotype was assessed by fluorescence-activated cell sorting, population doubling time was calculated and the multilineage differentiation assay was performed. Results: We have isolated multiple cell types from postnatal tissues. Namely, placental mesenchymal stromal cells from placenta chorionic disc, chorionic membrane mesenchymal stromal cells (ChM-MSC) from free chorionic membrane, umbilical cord MSC (UC-MSC) from whole umbilical cord, human umbilical vein endothelial cells (HUVEC) from umbilical vein, amniotic epithelial cells (AEC) and amniotic MSC (AMSC) from amniotic membrane. All isolated cell types displayed high proliferation rate together with the typical MSC phenotype: CD73+CD90+CD105+CD146+CD166+CD34-CD45-HLA-DR-. HUVEC constitutively expressed key markers CD31 and CD309. Most MSC and AEC were capable of osteogenic and adipogenic differentiation. Conclusion: We have shown that a wide variety of cell types can be easily isolated from extra-embryonic tissues and expanded ex vivo for regenerative medicine applications. These cells possess typical MSC properties and can be considered an alternative for adult MSC obtained from bone marrow or fat, especially for allogeneic use.
APA, Harvard, Vancouver, ISO, and other styles
37

Khanova, M. Yu, E. A. Velikanova, T. V. Glushkova, and V. G. Matveeva. "Development of personalized cell-populated vascular graft in vitro." Complex Issues of Cardiovascular Diseases 10, no. 2 (September 2, 2021): 89–93. http://dx.doi.org/10.17802/2306-1278-2021-10-2s-89-93.

Full text
Abstract:
Aim. To create a personalized cell-populated small-diameter vascular prosthesis in a pulsating bioreactor.Methods. Tubular grafts were made by electrospinning from mixtures of biodegradable polymers, poly (3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) and poly(εcaprolactone) (PCL). The inner surface is modified with fibrin. Tubular scaffolds were colonized with cultured colony-forming endothelial cells and grown under static conditions for 2 days. Then, the cell-populated prostheses continued to be cultivated for 5 days in a pulsating bioreactor system with a final shear stress of 2.85 dynes/cm².Results. The advantages of the cultivation of cell-populated vascular prostheses in a pulsating bioreactor have been revealed. The selected mode of cultivation of cellpopulated vascular prostheses under conditions of a pulsating flow with a shear stress of 2.85 dynes/cm² did not have a damaging effect on the integrity of the endothelial monolayer. Moving unidirectional mechanical stimuli of chaotic orientation fibers of F-actin changed to a predominant orientation in the direction of flow, and also increased the expression of F-actin, Talin focal adhesion protein, and specific endothelial markers CD309, CD31, vWF.Conclusion. The creation of a personalized cell-populated small-diameter vascular prosthesis with a functional endothelial monolayer is possible due to the use of autologous endothelial cells, autologous fibrin, and cultivation under conditions of a pulsating flow.
APA, Harvard, Vancouver, ISO, and other styles
38

Pulito-Cueto, Verónica, Sara Remuzgo-Martínez, Fernanda Genre, Belén Atienza-Mateo, Víctor M. Mora-Cuesta, David Iturbe-Fernández, Leticia Lera-Gómez, et al. "Endothelial Progenitor Cells: Relevant Players in the Vasculopathy and Lung Fibrosis Associated with the Presence of Interstitial Lung Disease in Systemic Sclerosis Patients." Biomedicines 9, no. 7 (July 20, 2021): 847. http://dx.doi.org/10.3390/biomedicines9070847.

Full text
Abstract:
Endothelial progenitor cells (EPC), which are key effectors in the physiologic vascular network, have been described as relevant players in autoimmune diseases. We previously showed that EPC frequency may help to identify the presence of interstitial lung disease (ILD) in rheumatoid arthritis patients. Given that ILD constitutes the main cause of mortality in systemic sclerosis (SSc) patients, we aimed to determine the EPC contribution to the pathogenic processes of vasculopathy and lung fibrosis in SSc-ILD+. EPC quantification was performed by flow cytometry on blood from 83 individuals: 21 SSc-ILD+ patients and subjects from comparative groups (20 SSc-ILD− and 21 idiopathic pulmonary fibrosis (IPF) patients and 21 healthy controls (HC)). EPC were considered as CD34+, CD45low, CD309+, and CD133+. A significant increase in EPC frequency was found in SSc-ILD+ patients when compared to HC (p < 0.001). SSc-ILD+ patients exhibited a higher EPC frequency than SSc-ILD− patients (p = 0.012), whereas it was markedly reduced compared to IPF patients (p < 0.001). EPC frequency was higher in males (p = 0.04) and negatively correlated to SSc duration (p = 0.04) in SSc-ILD+ patients. Our results indicate a role of EPC in the processes of vasculopathy and lung fibrosis in SSc-ILD+. EPC frequency may be considered as a biomarker of ILD in SSc patients.
APA, Harvard, Vancouver, ISO, and other styles
39

Pulito-Cueto, V., S. Remuzgo-Martínez, F. Genre, V. M. Mora-Cuesta, D. Iturbe Fernández, S. Fernández-Rozas, L. Lera-Gómez, et al. "SAT0014 ENDOTHELIAL PROGENITOR CELLS: ROLE IN ENDOTHELIAL DAMAGE OF INTERSTITIAL LUNG DISEASE ASSOCIATED TO RHEUMATOID ARTHRITIS." Annals of the Rheumatic Diseases 79, Suppl 1 (June 2020): 937.1–937. http://dx.doi.org/10.1136/annrheumdis-2020-eular.3228.

Full text
Abstract:
Background:Interstitial lung disease (ILD) is one of the most significant comorbidities of rheumatoid arthritis (RA), increasing the mortality in these patients [1,2]. Although the pathogenesis of ILD associated to RA (RA-ILD+) remains poorly defined [1], it is known that vascular tissue plays a crucial role in lung physiology [3]. In this context, a population of cells termed endothelial progenitor cells (EPC) are involved in vasculogenesis and endothelial tissue repair [4]. Previous reports suggest the implication of EPC in different conditions such as RA and idiopathic pulmonary fibrosis (IPF), the most common and destructive ILD [5,6]. Nevertheless, little is known about their specific role in RA-ILD+.Objectives:The purpose of this study was to shed light on the potential role of EPC in endothelial damage in RA-ILD+.Methods:Peripheral venous blood was collected from a total of 68 individuals (18 with RA-ILD+, 17 with RA-ILD-, 19 with IPF and 14 healthy controls). All subjects were recruited from the Rheumatology and Pneumology departments of Hospital Universitario Marqués de Valdecilla, Santander, Spain. Quantification of EPC was analyzed by the expression of surface antigens by flow cytometry. The combination of antibodies against the stem cell marker CD34, the immature progenitor marker CD133, the endothelial marker VEGF receptor 2 (CD309) and the common leukocyte antigen CD45 was used. EPC were considered as CD34+, CD45Low, CD309+and CD133+. All statistical analyses were performed using Prism software 5 (GraphPad).Results:EPC frequency was significantly increased in patients with RA-ILD+, RA-ILD-and IPF compared to controls (p=0.001, p=0.002, p< 0.0001, respectively). Nevertheless, patients with RA, both RA-ILD+and RA-ILD-, showed a lower frequency of EPC than those with IPF (p= 0.048, p= 0.006, respectively).Conclusion:Our results provide evidence for a potential role of EPC as a reparative compensatory mechanism related to endothelial damage in RA-ILD+, RA-ILD-and IPF patients. Interestingly, EPC frequency may help to establish a differential diagnostic between patients with IPF and those who have an underlying autoimmune disease (RA-ILD+).References:[1] J Clin Med 2019; 8: 2038;[2] Arthritis Rheumatol 2015; 67: 28-38;[3] Nat Protoc 2015; 10: 1697-1708;[4] Science 1997; 275: 964-966;[5] Rheumatology (Oxford) 2012; 51: 1775-1784;[6] Angiogenesis 2013; 16: 147-157.Acknowledgments:Personal funds, VP-C: PREVAL18/01 (IDIVAL); SR-M: RD16/0012/0009 (ISCIII-ERDF); LL-G: PI18/00042 (ISCIII-ERDF); RL-M: Miguel Servet type I CP16/00033 (ISCIII-ESF).Disclosure of Interests:Verónica Pulito-Cueto: None declared, Sara Remuzgo-Martínez: None declared, Fernanda Genre: None declared, Victor Manuel Mora-Cuesta: None declared, David Iturbe Fernández: None declared, Sonia Fernández-Rozas: None declared, Leticia Lera-Gómez: None declared, Pilar Alonso Lecue: None declared, Javier Rodriguez Carrio: None declared, Belén Atienza-Mateo: None declared, Virginia Portilla: None declared, David Merino: None declared, Ricardo Blanco Grant/research support from: AbbVie, MSD, Roche, Consultant of: Abbvie, Eli Lilly, Pfizer, Roche, Bristol-Myers, Janssen, UCB Pharma and MSD, Speakers bureau: Abbvie, Eli Lilly, Pfizer, Roche, Bristol-Myers, Janssen, UCB Pharma. MSD, Alfonso Corrales Speakers bureau: Abbvie, Jose Manuel Cifrián-Martínez: None declared, Raquel López-Mejías: None declared, Miguel A González-Gay Grant/research support from: Pfizer, Abbvie, MSD, Speakers bureau: Pfizer, Abbvie, MSD
APA, Harvard, Vancouver, ISO, and other styles
40

Franke, Anja C., Daniel Jung, and Thomas M. Ellis. "Characterization of the CD30L Binding Domain on the Human CD30 Molecule Using Anti-CD30 Antibodies." Hybridoma 19, no. 1 (February 2000): 43–48. http://dx.doi.org/10.1089/027245700315789.

Full text
APA, Harvard, Vancouver, ISO, and other styles
41

CHAKRABARTY, S., M. NAGATA, H. YASUDA, L. WEN, M. NAKAYAMA, S. A. CHOWDHURY, K. YAMADA, et al. "Critical roles of CD30/CD30L interactions in murine autoimmune diabetes." Clinical & Experimental Immunology 133, no. 3 (August 19, 2003): 318–25. http://dx.doi.org/10.1046/j.1365-2249.2003.02223.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
42

Lesteven, Elodie, Doriane Pognant, Valerie Vanneaux, Jérome Larghero, Olivier Feraud, Annelise Bennaceur-Griscelli, Emmanuelle Verger, et al. "Impact of the JAK2V617F Mutation on the Hemato-Endothelial Differentiation in an Induced Pluripotent Stem Cells (iPSC) Model." Blood 124, no. 21 (December 6, 2014): 4570. http://dx.doi.org/10.1182/blood.v124.21.4570.4570.

Full text
Abstract:
Abstract Background: Philadelphia-negative Myeloproliferative neoplasms (MPNs) are clonal disorders characterized by the acquisition of genetic alterations, the most frequent being the JAK2V617F mutation. Clinical complications include thrombo-hemorrhagic events and progression to either myelofibrosis or acute leukemia. MPNs are also associated with extramedullary hematopoiesis and increased vascularity in the spleen and bone marrow. Such neo-angiogenesis has been reported to involve the development of new microvessels through the local expansion of endothelial cells (ECs). Though JAK2V617F mutation was detected in ECs of hepatic venules of MPN patients with Budd-Chiari syndrome and in ECs derived from splenic capillaries, the presence of JAK2V617F in the endothelial lineage remains debatable and demonstration of hematopoietic and endothelial differentiation from progenitor cells harboring this mutation is lacking. We used a JAK2V617F iPSC line to assess the impact of this mutation on hematopoietic and endothelial differentiation. Methods: Hematopoietic and endothelial differentiation of an iPSC line harboring heterozygous JAK2V617F mutation were performed in two protocols: after derivation of embryoid bodies or with a MatrigelTMmatrix based protocol. Endothelial differentiation was performed after CD34+ or CD144+ cell sorting in classical endothelial conditions. Cells collected after differentiation in endothelial condition were evaluated for endothelial lineage surface markers, von Willebrand factor (vWF) expression, response to TNFα, and microvessel formation. Hematopoietic differentiation was assessed by flow cytometry and by clonogenic assay in methylcellulose with or without rh-EPO and rh-TPO. Results: After 6 days of differentiation in both protocols the emergence of CD34+ cells harboring the characteristics of an hemato-endothelial bipotent stem cell was observed: the phenotype of these cells was CD34+/CD90hi/CD38neg with partial expression of hematopoietic stem cell (HSC) markers like CD49f, CD117 (c-kit receptor), CXCR-4 but also of endothelial markers including CD31 (PECAM-1), CD309 (KDR) and CD144 (VE-cadherin); markers of hematopoietic commitment (CD43 or CD135) were absent. When transferred in an endothelial medium, CD34+ or CD144+ sorted derived cells were able to produce endothelial cells expressing CD54 (ICAM-1), CD31, CD144 and CD309, whereas the same cells produced the different hematopoietic lineages when maintained in hematopoietic differentiation protocol. PCR analyses confirmed that both cell types (endothelial and hematopoietic) harbored the JAK2V617F mutation and that the JAK2gene was expressed. Functionally, JAK2V617F-mutated endothelial cells derived from iPSCs expressed vWF in Weibel Palade bodies, responded normally to TNFα with the induction of CD106 (VCAM-1) expression and were able to generate micro-vessels on a Matrigel layer. Taken together these features indicate that the presence of the JAK2V617F mutation does not hamper endothelial specification or endothelial cells functionality. In contrast, the hematopoietic cells derived from the same JAK2V617F iPSC presented with functional abnormalities reminiscent of MPN such as cytokine-independent growth of erythroid and megakaryocytic colonies. Conclusion: This iPSC model provides new evidence for the possibility to generate a bipotent stem cell, able to produce both functional endothelial and hematopoietic lineages harboring the JAK2V617F mutation, from a common JAK2-mutated pluripotent stem cell. Our iPSC differentiation protocol also provides a new model to study the impact of the presence of JAK2V617F in ECs and could represent an effective tool for the screening of molecules antagonizing the pro-thrombotic or pro-hemorrhagic events in a JAK2V617F context. Disclosures No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
43

Jarajapu, Yagna PR, Sergio Caballero, Li Liu, Vinayak Shenoy, Michael J. Katovich, Mohan K. Raizada, and Maria B. Grant. "Activation of the Protective Arm of Renin Angiotensin System (RAS) Corrects the Reparative Dysfunction of Diabetic CD34+ Cells." Blood 116, no. 21 (November 19, 2010): 2637. http://dx.doi.org/10.1182/blood.v116.21.2637.2637.

Full text
Abstract:
Abstract Abstract 2637 Purpose: RAS plays a vital role in regulating many physiological processes of the vascular system. Angiotensin II (Ang II), a product of angiotensin converting enzyme (ACE), mediates its effects through activation of either the AT1 receptor – to induce vasoconstriction, proliferation, fibrosis, and inflammation – or the AT2 receptor to promote NO generation. The protective arm of RAS involves ACE2, which produces angiotensin-(1-7) [Ang-(1-7)]. Ang-(1-7) activates the MAS receptor to promote vascular health. Because diabetic endothelial progenitor cells are dysfunctional and this limits their utility in autologous cell therapy, we asked whether angiotensin (Ang)-(1-7) could restore the vasoreparative function of diabetic CD34+cells. Methods: Healthy nondiabetic (ND) and diabetic (D) Lin−CD45midCD34+ cells were obtained from peripheral blood mononuclear cells (PB-MNCs) by FACS. The effect of Ang-(1-7) on migration, proliferation, NO bioavailability, reactive oxygen species (ROS) levels and NADPH oxidase activity were evaluated in ND- and D-CD34+ cells. The effect of Ang-(1-7) on the formation of ECFCs from ND- and D-MNCs was evaluated. Ang-(1-7) production by cells was analyzed and the expression of ACE2 and Mas-receptor were assessed by real-time PCR and flow cytometry. Effects of ACE2 activators XNT and DIZE in CD34+ cells were also evaluated. D-CD34+ cells were genetically modified to overexpress Ang-(1-7) by lentiviral Ang-(1-7)-fusion transgene and their function was evaluated in vitro. The effect of transduction on the surface expression of CD133, CD34 and CD309 was assessed. In vivo homing function was assessed in a mouse model of ischemia-reperfusion (I/R). After one week of I/R insult, when retinal capillary damage was appreciable, CD34 cells were intravitreally injected. Neural retinas were harvested after 48 hours and human cells within the mouse vasculature were localized by immunohistochemistry. Results: Migration to SDF1- and VEGF-were impaired in D-CD34+ cells. In contrast, Ang-(1-7)-induced migration in both D-CD34+ and ND-CD34+cells was dependent on Mas receptor expression and eNOS. ROS levels and NADPH oxidase activity were reduced and proliferation and NO bioavailability were restored in D-CD34+ cells by Ang-(1-7). ECFCs from D-MNCs were appeared only in the presence of Ang-(1-7). Migration, NO release and Ang-(1-7) release by ACE2 activators, XNT and DIZE, were significantly decreased in D-CD34+ cells. Ang-(1-7) release and ACE2 expression were decreased in D-cells while Mas-receptor expression was similar that in ND-cells. Ang-(1-7) gene-modified cells showed reduced ROS levels, increased NO bioavailability, enhanced migration to SDF-1 and proliferation. Lentiviral transduction did not alter surface expression of CD133, CD34 and CD309. Ang-(1-7)-overexpression restored the homing efficiency of D-CD34+ cells similar to that of ND-CD34+ cells in vivo. Conclusions: Ang-(1-7) stimulates the vasoreparative functions in CD34+ cells. Ang-(1-7) bypasses the reduced ACE2 seen in diabetic CD34+ cells and restores the vasoreparative potential of these cells by decreasing oxidative stress and normalizing NO bioavailability. Pharmacological strategies that either increase ACE2 or Ang-(1-7) in diabetic CD34+ cells will improve their therapeutic utility for autologous cell therapy in treatment of diabetic complications. Disclosures: No relevant conflicts of interest to declare
APA, Harvard, Vancouver, ISO, and other styles
44

Huhn, Michael, Stephanie Sasse, Mehmet K. Tur, T. Schinköthe, Susanna M. Rybak, Andreas Engert, and Stefan Barth. "Human angiogenin fused to human CD30 ligand (Ang-CD30L) exhibits specific cytotoxicity against CD30-positive lymphoma." European Journal of Cancer 37 (September 2001): S12. http://dx.doi.org/10.1016/s0959-8049(01)80351-8.

Full text
APA, Harvard, Vancouver, ISO, and other styles
45

Bratseth, Vibeke, Gemma Chiva-Blanch, Rune Byrkjeland, Svein Solheim, Harald Arnesen, and Ingebjørg Seljeflot. "Elevated levels of circulating microvesicles in coronary artery disease patients with type 2 diabetes and albuminuria: Effects of exercise training." Diabetes and Vascular Disease Research 16, no. 5 (April 26, 2019): 431–39. http://dx.doi.org/10.1177/1479164119843094.

Full text
Abstract:
Objective:Circulating microvesicles, released from activated/apoptotic cells, are involved in vascular complications and may be looked upon as biomarkers. Albuminuria is characteristic of disease progression in type 2 diabetes mellitus. We aimed to investigate quantitative and qualitative differences of circulating microvesicles in type 2 diabetes mellitus with and without albuminuria and whether 12-month exercise training influenced expression of circulating microvesicles.Methods:Coronary artery disease patients with type 2 diabetes mellitus (n = 75), of which 25 had albuminuria, were included. Annexin V+(AV+) circulating microvesicles were analysed by flow cytometry in citrated plasma. The exercise volume was 150 min per week.Results:In albuminuria patients, circulating microvesicles from endothelial-(CD146+/CD62E+/AV+) and endothelial-progenitor-(CD309+/CD34+/AV+) cells were significantly higher compared to those without ( p ⩽ 0.01, both). Receiver operating characteristic curve analysis of the endothelial circulating microvesicles shows an area under the curve of 0.704 (95% confidence interval: 0.57–0.84; p = 0.004). Albuminuria patients had more circulating microvesicles derived from activated leukocytes and monocytes and monocytes carrying tissue factor (CD11b+/AV+, CD11b+/CD14+/AV+, CD142+/CD14+/AV+, respectively, p ⩽ 0.05, all) and higher number of circulating microvesicles from activated platelets (CD62P+/AV+). Within exercising patients, circulating microvesicles from progenitor cells increased ( p = 0.023), however, not significantly different from controls.Conclusion:Coronary artery disease patients with type 2 diabetes mellitus and albuminuria had elevated number of circulating microvesicles from activated blood and vascular cells, rendering them as potential predictors of disease severity. The circulating microvesicles were limitedly affected by long-term exercise training in our population.
APA, Harvard, Vancouver, ISO, and other styles
46

Rossi, Francesca Maria, Massimo Degan, Linda Mazzocut-Zecchin, Raffaele Di Francia, Donatella Aldinucci, Antonio Pinto, and Valter Gattei. "CD30L up-regulates CD30 and IL-4 expression by T cells." FEBS Letters 508, no. 3 (November 23, 2001): 418–22. http://dx.doi.org/10.1016/s0014-5793(01)03076-9.

Full text
APA, Harvard, Vancouver, ISO, and other styles
47

Guo, Feng, Zi-Xing Chen, Aining Sun, Peng Zhou, and Wenjuan Wang. "Differential Effects of TRAFs in the Activation of NF-KappB in Hodgkin’s Lymphoma Cells." Blood 112, no. 11 (November 16, 2008): 1458. http://dx.doi.org/10.1182/blood.v112.11.1458.1458.

Full text
Abstract:
Abstract CD30, a member of the TNF receptor (TNFR) superfamily, is a lymphocyte-specific receptor that is originally recognized as a surface molecule overexpressed on Hodgkin’s lymphoma cells. Engagement of CD30 with its ligand CD30L activates the nuclear factor kB (NF-kB), which are mediated by interactions with TNFR-associated factor (TRAFs), and supports proliferation of Hodgkin’s cells. However, the role of individual TRAFs in the CD30 signaling pathway in Hodgkin’s cells has not been fully addressed yet. In this study, we found that except TRAF3, all other TRAFs were expressed consistently in B cell-derived Hodgkin’s cell lines (L428 and KM-H2) either in mRNA or protein level. In L428 but not KM-H2 cells, the classical (p50-RelA) and alternative (p52-RelB) NF-kB activity were constitutively activated measured by Western Blotting and EMSA. To better understand the CD30 signaling properties in Hodgkin’s lymphoma cells, we silenced the expression of TRAFs individually by means of RNAi. Successful downregulation of TRAF1 protein expression led to the apoptosis of L428 cells, which was companied by the reduction of both classical and alternative NF-kB activity. Furthermore, the expression of targeting genes of NF-kB, such as A20, c-Flip, ICAM-1, and Cyclin D1 was also decreased. Using siRNA targeting TRAF2 expression, the classical NF-kB activity was reduced while the alternative NF-kB activity moderately induced especially upon CD30L treatment, which was companied by the induction of p100 processing and RelB nuclear localization. The survival rate was decreased when TRAF2 was knockdown. TRAF5 knockdown manifested similar results as that of TRAF2. These observations were only took place in L428 cells but not KM-H2 cells. In addition, the phosphorylation of the extracellular signal-regulated kinases (ERK) 1 and 2, and subsequent activation of JunB in Hodgkin’s cells were staying unchanged in individual TRAFs knockdown experiment. Taken together, the study here further investigates the features of CD30-TRAFs-NF-kB signaling pathway in Hodgkin’s lymphoma cells. At least TRAF1, 2, and 5 are involved in CD30 signaling pathway and protecting Hodgkin’s lymphoma cells from undergoing apoptosis. The interaction between TRAFs is necessary to facilitate the signaling and proper activation of classical NF-kB, which is responsible for the antiapoptotic effect of Hodgkin’s cells. Among TRAF1, 2, and 5, the redundant function is unlikely. Furthermore, the sequential activation of ERK and JunB is unlikely dependent on TRAFs-NF-kB pathway in Hodgkin’s lymphoma cells. Thus, these data reveal that the classical NF-kB activity downstream of CD30 signaling is TRAFs-dependent in Hodgkin’s lymphoma cells, which is regulated by the relative level of individual TRAF in a cell-specific manner.
APA, Harvard, Vancouver, ISO, and other styles
48

Rossi, Francesca Maria, Davide Rossi, Antonella Zucchetto, Riccardo Bomben, Michele Dal Bo, Dania Benedetti, Claudio Tripodo, et al. "CD49d Expression Identifies a Chronic Lymphocytic Leukemia (CLL) Subset with High Levels of Circulating CD34 +Cells Co-Expressing Endothelial Cell Markers." Blood 114, no. 22 (November 20, 2009): 2329. http://dx.doi.org/10.1182/blood.v114.22.2329.2329.

Full text
Abstract:
Abstract Abstract 2329 Poster Board II-306 Introduction: In chronic lymphocytic leukemia (CLL), CD49d, often in association with CD38, has been shown to mark a disease subset with poor prognosis. Functionally, both molecules act as counter-receptors for surface structures (i.e. VCAM-1/CD106 and CD31) usually expressed by the endothelial/stromal component of tumor micro-environment. We have recently identified a micro-environmental circuitry which involves CD38 triggering, and eventually determines an enrichment of the VCAM-1/CD106-expressing endothelial component detected in the context of CLL infiltrates found in bone marrow biopsies. Data was also provided that CD49d/VCAM-1 interactions are active in delivering pro-survival signals to CD49d-expressing CLL cells (Zucchetto et al, Cancer Res, 69, 4001, 2009). In this study, we investigated the amount of circulating progenitors with endothelial phenotype in CLL samples with different CD49d and CD38 expression levels. Methods: Peripheral blood (PB) samples from 91 CLL cases purposely selected with WBC>25,000/μl (B cells absolute lymphocyte count >10,000/μl) were evaluated by multiparametric flow cytometry for the absolute count of circulating CD34+ cells (ISHAGE protocol in single platform). Whenever possible (i.e. if a cluster of at least 100 CD34+ cells was detectable), a further characterization was performed (4-6 colours flow cytometry) for circulating endothelial cells (CEC), identified as a CD34+CD45low cell population co-expressing one of the following endothelial markers: CD309/VEGFR-2, CD144/VE-cadherin, CD106/VCAM-1 and CD146/Muc-18. CD49d and CD38 expression by CLL cells was considered positive if exceeding the standard cut-off value of 30% of positive cells. Results: PB absolute CD34+ cell counts were 7.5±7.5/μl in CD49d+ CLL (32 cases), vs. 3.3±2.7/μl in CD49d− CLL (59 cases; p=2.6×10−4), or 9.4±8.7/μl in CD49d+ CLL (30 cases) vs. 4.6±2.9/μl in CD49d− CLL (18 cases; p=0.004) when only cases phenotyped for CEC were considered. Furthermore, when samples were stratified also for CD38 expression, values of circulating CD34+ cells increased to 10.6±10.1/μl in CD38+CD49d+ CLL (11 cases) vs. 3.1±2.4/μl in CD38−CD49d− (51cases; p=1×10−5). Regarding the absolute quantification of CEC, a CD49d+ phenotype again marked the CLL subset with the highest CEC count, as identified by the expression of either the CD309/VEGFR-2 (CEC counts 1.7±2.3/μl in CD49d+ CLL vs. 0.5±0.5/μl CD49d− CLL; p=0.009) or the CD144/VE-cadherin (CEC counts 0.8±1/μl in CD49d+ CLL vs. 0.3±0.5/μl in CD49d− CLL; p=0.057) endothelial markers on CD34+CD45low cells. Notably, CEC from CD49d+ CLL expressed CD106/VCAM-1 in virtually all cells (1.6±2.4/μl), while the other marker of endothelial activation CD146/Muc-18 was detected in a fraction of CEC only (0.4±0.9/μl). Conclusions: CD49d and CD38 expression by CLL cells identify a disease subset with significantly higher number of both circulating CD34+ cells and CEC. This phenomenon could be explained considering several aspects: i) the sharing of common phenotypic markers between CLL cells and CD34+ progenitors, including CD38 and CD49d, which could be responsible for a displacement of CD34+ progenitors in the context of micro-environmental niches; ii) the known capacity of CLL cells, especially with a unmutated IGHV gene status and/or a CD38+CD49d+ phenotype to produce pro-angiogenic factors including Ang-2; iii) the rare PB cells expressing CD34 and CEC markers may represent CLL cell precursors with tumor-initiating cell features. Studies are currently ongoing to dissect among these hypotheses Disclosures: No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
49

Erkek, Esra Turan, Esra Nazligul, Meliha Nalcaci, Melih Aktan, and Mustafa Nuri Yenerel. "Circulating Endothelial Progenitor Cells and Their Relation to Thrombosis in Paroxysmal Nocturnal Hemoglobinuria." Blood 126, no. 23 (December 3, 2015): 4535. http://dx.doi.org/10.1182/blood.v126.23.4535.4535.

Full text
Abstract:
Abstract Introduction: Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired clonal hematopoietic stem cell disorder. Chronic intravascular hemolytic anemia, bone marrow failure and thrombophilia are the main clinical findings. Thrombosis is one of the most important cause of morbidity and mortality of this disease. Multiple factors are held responsible for thrombotic tendency in these patients. Endothelial progenitor cells (EPCs) originate from primitive hematopoietic stem cells and are able to turn into endothelial cells. There are extremely small numbers of EPCs in the circulation under normal conditions. The level of EPCs is considered to be indicative of restoration capacity in case of vascular disease and potential damage. Lower EPC levels are also considered as a risk factor in cardiovascular diseases. In this study, our aim was to investigate circulating EPCs in PNH and their relationship with thrombosis. Seventeen patients with PNH, 18 patients with aplastic anemia and 10 healthy volunteers were included in the study. CD309, CD133 and CD34 antibodies were used in order to determine circulating EPCs by flow cytometry and cells which expressed all three antibodies were analyzed as EPC. Prepared samples were read using a prepared list mode software for endothelial progenitor cells on FACS Diva software in BD FACS Canto II device with 6 color lasers and a total of 1,000,000 cells per analysis were evaluated. EPC levels were compared between untreated PNH patients and who were on eculizumab therapy. Statistical analysis was performed using SPSS 22.0 software. The distribution of variables was evaluated by Kolmogorov-Smirnov test, the analysis of quantitative data was evaluated by ANOVA, Kruskal-Wallis, Mann-Whitney U tests and the analysis of the qualitative data was evaluated by chi-square test. Findings and Discussion: The thrombotic complications were observed in five PNH patients. All of these patients had a history of portal vein thrombosis. One of them also had a history of peripheral arterial disease and amputation related to this. There was not a significant difference in EPC levels between patients with and without a history of thrombosis (p>0,05). We also did not find any significant difference between levels of EPC's in PNH groups with or without eculizumab therapy (p˃0,05). There was no significant difference in levels of EPC between aplastic anemia and PNH groups (p ˃ 0,05). However, we found a significant positive correlation between the levels of EPC and LDH in multivariate analysis (p < 0,05). This finding suggests that hemolysis causes vascular endothelium and promotes new blood vessel formations. Increased EPCs in PNH might be an indirect indicator for vascular endothelium damage in PNH. Table. General Features and Rates of EPC of PNH, AA, Healthy Volunteers Groups Aplastic Anemia group PNH group Control group p Age mean±s.smedian (min-max) 40.0±14.7 37.5 (20.0-67.0) 41.9±13.9 43.0 (19.0-78.0) 29.3±3.5 29.5(24.0-34.0) 0.047 Sex Female n-% Male n-% 7 38,9% 11 61.1% 9 52.9% 8 47.1% 5 50% 5 50% 0.687 EPC(%) mean±s.s median (min-max) 0.2% 0.2% 0.1% (0.0-0.6%) 0.3%±0.3% 0.1% (0.0-0.9%) 0.1%±0.0% %0,0(%0,0-0,2) 0.393 All Events (x1000) mean±s.s median (min-max) 617±172* 565 (360-914) 588±255* 471 (250-1000) 878±143 950(655-1000) 0.003 CD309 and CD34 mean±s.s median (min-max) 0.003±0.002 0.002 (0.001-0.007) 0.005±0.004 0.000-0.011) 0.001±0.001 0.001 (0.000-0.002) 0.009 CD133 mean±s.s median (min-max) 45.8±36.3 58.3 (0.0-88.2) 45.8±39.6 60 (0.0-94.7) 42.0±19.4 46.4 (11.1-81.3) 0.867 ANOVA / Kruskal-Wallis / Mann-Whitney U test / Chi-square test *The difference with the control group, p <0.05 EPCs: Endothelial progenitor cells Disclosures Yenerel: Alexion: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau.
APA, Harvard, Vancouver, ISO, and other styles
50

Głowińska-Olszewska, Barbara, Marcin Moniuszko, Andrzej Hryniewicz, Marta Jeznach, Małgorzata Rusak, Milena Dąbrowska, Włodzimierz Łuczyński, Anna Bodzenta-Łukaszyk, and Artur Bossowski. "Relationship between circulating endothelial progenitor cells and endothelial dysfunction in children with type 1 diabetes: a novel paradigm of early atherosclerosis in high-risk young patients." European Journal of Endocrinology 168, no. 2 (February 2013): 153–61. http://dx.doi.org/10.1530/eje-12-0857.

Full text
Abstract:
ObjectiveThe low number of circulating endothelial progenitor cells (EPCs) has emerged as a biomarker of cardiovascular (CV) risk in adults. Data regarding EPCs in paediatric populations with CV risk factors are limited. The aim of the study was to estimate the EPC number and its relationship with vascular function and structure in children with type 1 diabetes mellitus (T1DM).Design and methodsWe performed a comparative analysis of 52 children with T1DM (mean age 14.5 years; diabetes duration, 6.0 years; HbA1c level, 8.5%) and 36 healthy age- and gender-matched control children. EPCs were identified and analysed by flow cytometry with the use of MABs directed against CD34, CD144 (VE-cadherin) and CD309 (VEGFR-2). sICAM-1, hsCRP, thrombomodulin and adiponectin levels were also assessed. We evaluated vascular function (flow-mediated dilation (FMD)) and structure (carotid intima–media thickness (IMT)) ultrasonographically.ResultsFrequencies of CD34+ cells were similar in both groups (P=0.30). In contrast, frequencies of CD34+VE-cadherin+ cells were significantly higher in diabetic children compared with the healthy group (P=0.003). Similarly, diabetic patients tended to present with higher frequencies of CD34+VEGFR+ cells (P=0.06). FMD was lower (6.9 vs 10.5%, P=0.002) and IMT was higher (0.50 vs 0.44 mm, P=0.0006) in diabetic children. We demonstrated a significant relationship between CD34+VEGFR-2+ cells and BMI (r=0.3, P=0.014), HDL (r=−0.27, P=0.04), sICAM-1 (r=0.47, P=0.023) and FMD (r=−0.45, P<0.001). Similarly, frequencies of CD34+VE-cadherin+ cells were significantly correlated with BMI (r=0.32, P=0.02) and FMD (r=−0.31, P=0.03).ConclusionsWe demonstrated here that increased frequencies of EPCs observed in diabetic children are negatively correlated with endothelial function. Further studies are warranted to assess whether this phenomenon might result from effective mobilisation of EPCs in order to repair damaged endothelium in children at increased risk for atherosclerosis.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography