Regulation of Constitutive Tip110 Expression in Human Cord Blood CD34+Cells Through Selective Usage of the Proximal and Distal Polyadenylation Sites Within the 3′Untranslated Region

2018 ◽  
Vol 27 (8) ◽  
pp. 566-576
Author(s):  
Ying Liu ◽  
Xinxin Huang ◽  
Khalid A. Timani ◽  
Hal E. Broxmeyer ◽  
Johnny J. He
Blood ◽  
2000 ◽  
Vol 96 (13) ◽  
pp. 4169-4177 ◽  
Author(s):  
Adeline Lepage ◽  
Marylène Leboeuf ◽  
Jean-Pierre Cazenave ◽  
Corinne de la Salle ◽  
François Lanza ◽  
...  

Abstract Megakaryocytopoiesis is a complex multistep process involving cell division, endoreplication, and maturation and resulting in the release of platelets into the blood circulation. Megakaryocytes (MK) progressively express lineage-restricted proteins, some of which play essential roles in platelet physiology. Glycoprotein (GP)Ib-V-IX (CD42) and GPIIb (CD41) are examples of MK-specific proteins having receptor properties essential for platelet adhesion and aggregation. This study defined the progressive expression of the GPIb-V-IX complex during in vitro MK maturation and compared it to that of GPIIb, an early MK marker. Human cord blood CD34+ progenitor cells were cultured in the presence of cytokines inducing megakaryocytic differentiation. GPIb-V-IX expression appeared at day 3 of culture and was strictly dependent on MK cytokine induction, whereas GPIIb was already present in immature CD34+ cells. Analysis by flow cytometry and of the messenger RNA level both showed that GPV appeared 1 day later than GPIb-IX. Microscopy studies confirmed the late appearance of GPV, which was principally localized in the cytoplasm when GPIb-IX was found on the cell surface, suggesting a delayed program of GPV synthesis and trafficking. Cell sorting studies revealed that the CD41+GPV+ population contained 4N and 8N cells at day 7, and was less effective than CD41+GPV− cells in generating burst-forming units of erythrocytes or MK colonies. This study shows that the subunits of the GPIb-V-IX complex represent unique surface markers of MK maturation. The genes coding for GPIb-IX and GPV are useful tools to study megakaryocytopoiesis and for tissue-specific or conditional expression in mature MK and platelets.


2015 ◽  
Vol 6 (1) ◽  
Author(s):  
Yuxia Yang ◽  
Saifeng Wang ◽  
Zhenchuan Miao ◽  
Wei Ma ◽  
Yanju Zhang ◽  
...  

2001 ◽  
Vol 16 (1) ◽  
pp. 20 ◽  
Author(s):  
Ju Young Seoh ◽  
Hae Young Park ◽  
Wha Soon Chung ◽  
Seung Cheol Kim ◽  
Myong Joon Hahn ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3451-3451
Author(s):  
Jong-Ho Won ◽  
Hee-Jeong Cheong ◽  
Sook-Ja Kim ◽  
Sang-Byung Bae ◽  
Chan-Kyu Kim ◽  
...  

Abstract The anemia of chronic disease-which encompasses inflammation, infection, tissue injury, and conditions associated with the release of proinflammatory cytokines (such as cancer)- is one of the most common forms of anemia seen clinically. Symptomatic anemia requires treatment. The two major forms of treatment are transfusions and erythropoietin. Arsenic trioxide (As2O3) used to treat human diseases for centuries in traditional Chinese medicine. Our recent studies suggest that low dose of As2O3 induces erythroid differentiation of K562 human leukemic cells and high dose of As2O3 induce apoptosis. In this study, we have investigated in vitro effect of As2O3 on the erythroid differentiation and it could inhibit TNF-α induced suppression of erythroid differentiation of human cord blood CD34+ cells. Expression of glycophorin A was 35.94 ± 7.94% after 7 days culture of human cord blood CD34+ cells and was decreased to 17.63 ± 7.33% when culture of human cord blood CD34+ cells with 100ng/mL of TNF-α. Expression of glycophorin A was increased in dose dependent manner after 7 days treatment with As2O3 and As2O3 increased percentage of glycophorin A in culture with TNF-α compared to TNF-α alone. The results of colony assay of CFU-MIX and BFU-E after culture with various conditions revealed similar patterns with expression of glycophorin A. These results suggest that As2O3 induces erythroid differentiation of human cord blood CD34+ cells and can reverse TNF-α induced suppression of erythroid differentiation of human cord blood CD34+ cells. The BFU-E colony assay of the human cord blood CD34+ cells after culture with TNF-α or/and Arsenic trioxide. The BFU-E colony assay of the human cord blood CD34+ cells after culture with TNF-α or/and Arsenic trioxide.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1329-1329
Author(s):  
Aleksandra Rizo ◽  
Edo Vellenga ◽  
Gerald de Haan ◽  
Jan Jacob Schuringa

Abstract Hematopoietic stem cells (HSCs) are able to self-renew and differentiate into cells of all hematopoietic lineages. Because of this unique property, they are used for HSC transplantations and could serve as a potential source of cells for future gene therapy. However, the difficulty to expand or even maintain HSCs ex vivo has been a major limitation for their clinical applications. Here, we report that overexpression of the Polycomb group gene Bmi-1 in human cord blood-derived HSCs can potentially overcome this limitation as stem/progenitor cells could be maintained in liquid culture conditions for over 16 weeks. In mouse studies, it has been reported that increased expression of Bmi-1 promotes HSC self-renewal, while loss-of-function analysis revealed that Bmi-1 is implicated in maintenance of the hematopoietic stem cells (HSC). In a clinically more relevant model, using human cord blood CD34+ cells, we have established a long-term ex-vivo expansion method by stable overexpression of the Bmi-1 gene. Bmi-1-transduced cells proliferated in liquid cultures supplemented with 20% serum, SCF, TPO, Flt3 ligand, IL3 and IL6 for more than 4 months, with a cumulative cell expansion of more then 2×105-fold. The cells remained cytokine-dependent, while about 4% continued to express CD34 for over 20 weeks of culture. The cultured cells retained their progenitor activity throughout the long-term expansion protocol. The colony-forming units (CFUs) were present at a frequency of ~ 30 colonies per 10 000 cells 16 weeks after culture and consisted of CFU-GM, BFU-E and high numbers of CFU-GEMM type progenitors. After plating the transduced cells in co-cultures with the stromal cell line MS5, Bmi-1 cells showed a proliferative advantage as compared to control cells, with a cumulative cell expansion of 44,9 fold. The non-adherent cells from the co-cultures gave rise to higher numbers of colonies of all types (~70 colonies/10.000 cells) after 4 weeks of co-culture. The LTC-IC frequencies were 5-fold higher in the Bmi-1-transduced cells compared to control cells (1/361 v.s. 1/2077, respectively). Further studies will be focused on in-vivo transplantation of the long-term cultured cells in NOD/SCID mice to test their repopulating capacity. In conclusion, our data implicate Bmi-1 as an important modulator of human HSC self-renewal and suggest that it can be a potential target for therapeutic manipulation of human HSCs.


2001 ◽  
Vol 113 (2) ◽  
pp. 470-478 ◽  
Author(s):  
Kyung-Ha Ryu ◽  
Susan Chun ◽  
Steve Carbonierre ◽  
Seock-Ah Im ◽  
Hyung-Lae Kim ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2100-2100
Author(s):  
Yasuo Nagasawa ◽  
Brent Wood ◽  
Cynthia L. Nourigat ◽  
Thalia Papayannopoulou ◽  
C. Anthony Blau

Abstract Cell growth switches are engineered signaling molecules that can trigger cell growth in response to artificial ligands such as chemical inducers of dimerization (CIDs). We have previously shown that a cell growth switch comprised of the intracellular portion of the thrombopoietin receptor, Mpl, allows for the CID-dependent, in vivo expansion of genetically modified primary hematopoietic cells in mouse and dog models. Here we report the application of this approach to the in vivo expansion of genetically modified primary human hematopoietic cells using an immune deficient mouse model. A lentivirus vector encoding a CID-activatible deriviative of Mpl (F36VMpl) and a green fluorescent protein (GFP) reporter was used to transduce human cord blood CD34+ cells. Transduced human cord blood CD34+ cells expanded 347–495 fold in cultures containing no added growth factors and 100 nM of AP20187. We proceeded to test CID-responsiveness following transplantation into NOD-SCID-beta 2 microglobulin null mice. Since significant human red cell engraftment persists for only a few weeks post transplantation in this model, mice were evaluated at 3 weeks post transplantation, following a 2 week course of treatment with either CID (AP20187 10 mg/kg/day) or control vehicle alone (without CID). In 2 experiments totalling 42 mice, CID-administration resulted in a significant rise in GFP positive human cells, with the predominant response occuring among human erythroid cells (exp.1: 4.0 fold, P = 0.0003, exp.2: 12.7 fold, P = 0.038). An increase in transduced human erythroid progenitor cells was observed in the spleens (but not the femurs) of CID treated mice. Effects on other hematopoietic lineages were minor and variable. The effect of CID treatment was no longer evident five weeks after the last dose. The restriction of CID-induced cell growth to the erythroid lineage may make this approach well suited for gene therapy applications in sickle cell anemia and beta thalassemia.


Sign in / Sign up

Export Citation Format

Share Document