An expansion phase precedes terminal erythroid differentiation of hematopoietic progenitor cells from cord blood in vitro and is associated with up-regulation of cyclin E and cyclin-dependent kinase 2

Blood ◽  
2000 ◽  
Vol 96 (12) ◽  
pp. 3985-3987 ◽  
Author(s):  
Mu-Shui Dai ◽  
Charlie R. Mantel ◽  
Zhen-Biao Xia ◽  
Hal E. Broxmeyer ◽  
Li Lu

The dynamics of cell cycle regulation were investigated during in vitro erythroid proliferation and differentiation of CD34+cord blood cells. An unusual cell cycle profile with a majority of cells in S phase (70.2%) and minority of cells in G1 phase (27.4%) was observed in burst-forming unit-erythrocytes (BFU-E)–derived erythroblasts from a 7-day culture of CD34+ cells stimulated with interleukin 3 (IL-3), granulocyte-macrophage colony-stimulating factor (GM-CSF), Steel factor, and Epo. Terminal erythroid differentiation was accompanied by a rapid increase of G0/G1 phase cells. Expression of cyclin E and cyclin-dependent kinase 2 (cdk2) correlated with the proportion of S phase cells. Cyclin D3 was moderately up-regulated during the proliferation phase, and both cyclin E and D3 were rapidly down-regulated during terminal differentiation. This suggests that the high proliferation potential of erythroblasts is associated with temporal up-regulation of cyclin E and cdk2.

Blood ◽  
2000 ◽  
Vol 96 (12) ◽  
pp. 3985-3987 ◽  
Author(s):  
Mu-Shui Dai ◽  
Charlie R. Mantel ◽  
Zhen-Biao Xia ◽  
Hal E. Broxmeyer ◽  
Li Lu

Abstract The dynamics of cell cycle regulation were investigated during in vitro erythroid proliferation and differentiation of CD34+cord blood cells. An unusual cell cycle profile with a majority of cells in S phase (70.2%) and minority of cells in G1 phase (27.4%) was observed in burst-forming unit-erythrocytes (BFU-E)–derived erythroblasts from a 7-day culture of CD34+ cells stimulated with interleukin 3 (IL-3), granulocyte-macrophage colony-stimulating factor (GM-CSF), Steel factor, and Epo. Terminal erythroid differentiation was accompanied by a rapid increase of G0/G1 phase cells. Expression of cyclin E and cyclin-dependent kinase 2 (cdk2) correlated with the proportion of S phase cells. Cyclin D3 was moderately up-regulated during the proliferation phase, and both cyclin E and D3 were rapidly down-regulated during terminal differentiation. This suggests that the high proliferation potential of erythroblasts is associated with temporal up-regulation of cyclin E and cdk2.


2004 ◽  
Vol 24 (13) ◽  
pp. 6058-6066 ◽  
Author(s):  
Xin-Hua Zhu ◽  
Hoang Nguyen ◽  
H. Dorota Halicka ◽  
Frank Traganos ◽  
Andrew Koff

ABSTRACT Ubiquitin-dependent proteolysis makes a major contribution to decreasing the levels of p27. Ubiquitin-dependent proteolysis of p27kip1 is growth and cell cycle regulated in two ways: first, skp2, a component of the E3-ubiquitin ligase, is growth regulated, and second, a kinase must phosphorylate the threonine-187 position on p27 so that it can be recognized by skp2. In vitro, p27 is phosphorylated by cyclin E- and cyclin A-associated cdk2 as well as by cyclin B1-cdk1. Having analyzed the effect of different cyclin-cyclin-dependent kinase complexes on ubiquitination of p27 in a reconstitution assay system, we now report a noncatalytic requirement for cyclin A-cdk2. Multiparameter flow cytometric analysis also indicates that p27 turnover correlates best with the onset of S phase, once the levels of cyclin A become nearly maximal. Finally, increasing the amount of both cyclin E-cdk2 and skp2 was less efficient at promoting p27 ubiquitination than was increasing the amount of cyclin A-cdk2 alone in extracts prepared from cultures of >93%-purified G1 cells. Together these lines of evidence suggest that cyclin A-cdk2 plays an ancillary noncatalytic role in the ubiquitination of p27 by the SCFskp2 complex.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2474-2474
Author(s):  
Piotr Smolewski ◽  
Agnieszka Janus ◽  
Barbara Cebula ◽  
Anna Linke ◽  
Krzysztof Jamroziak ◽  
...  

Abstract Background: Rapamycin (RAPA) is an inhibitor of mTOR kinase pathway. In vitro low doses of this agent induce cell cycle arrest in G1 phase, whereas higher concentrations of RAPA exert proapoptotic effects. Aim: We assessed cytotoxicity of RAPA alone or in combination with cytarabine (cytosine arabinoside, ARA-C) in acute myeloblastic leukemia (AML) cells and in normal lymphocytes obtained from 10 healthy volunteers. Methods: AML cells (in vitro HL-60 cell line and ex vivo leukemic cells) and phytohemaglutynin (PHA)-stimulated normal lymphocytes were treated for 24 – 48 h with 1 ng/ml RAPA alone or in combination with 50 nM cytarabine (Ara-C). Moreover, cells was pre-incubated with RAPA for 24 h and then Ara-C was added for the next 24 h. Untreated cultures and those treated with RAPA, Ara-C or PHA alone served as respective controls. The proapoptotic effect was assessed by Annexin V assay and presented as a percentage of Annexin-V-positive cells (apoptotic index; AI). Cell cycle was analyzed by DNA distribution in propydium iodide/RN-ase stained cells. Cyclin D3, A and E expression was also measured using flow cytometry. Results: Median AI induced in HL-60 cells after 24 h treatment with RAPA+Ara-C (30.1%) was significantly higher than induced by RAPA (7.2%) or Ara-C (18.5%) alone (p=0.002 and p=0.03, respectively). The RAPA+Ara-C combination exerted additive effect (combination index 0.87) in that model. Additional 24 hour pretreatment with RAPA further increased apoptosis (median AI 41.5%, vs. 10.9% after 48 h-RAPA alone). In contrast to leukemic cells, pretreatment of normal PHA-stimulated lymphocytes with RAPA caused their G1 phase cell cycle arrest, with significant decrease in cyclin D3 expression (vs. untreated cells - p<0.001). This resulted in prevention of Ara-C-induced cytotoxicity in healthy lymphocytes, when Ara-C was added for another 24 h. Importantly, that protective effect was reversible when RAPA-treated lymphocytes were rinsed and then cultured in fresh, RAPA-free medium for the next 24 h. In another set of experiments, cells from 12 de novo AML patients were treated with RAPA and Ara-C in above concentrations and time settings. RAPA and Ara-C were administrated to isolated peripheral blood mononuclear cells (PBMC). PBMC were immunophenotyped before and after treatment. Leukemic blasts were marked for individually chosen antigen, most characteristic for leukemic clone in particular patient. Normal CD3+ lymphocytes were also detected. Finally, Annexin V staining was performed. Based on that simultaneous three-color staining the proapoptotic effects of treatment could be measured by flow cytometry in both leukemic blasts and normal CD3+ cells. Thus, we found that pretreatment with RAPA protected majority of CD3+ cells (median of alive cells 85.5%) from Ara-C-induced apoptosis, whereas the leukemic blasts AI was higher than in samples treated with Ara-C. After Ara-C alone CD3+ rate decreased significantly (median 35.1%). Conclusions: Pretreatment with RAPA enhances cytotoxic effect of Ara-C on leukemic cells, but not on healthy lymphocytes. The phenomenon is probably due to reversible arrest of healthy cells in G1 phase of cell cycle by low doses of RAPA, what causes their transient resistance to proapoptotic action of cytostatic drugs. In contrast, the same RAPA doses selectively sensitizes leukemic cells to cytostatics. This suggests, that inhibition of mTOR kinase prior to cytostatics administration may result in selective anti-tumor treatment, with protection of normal cells.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2580-2580
Author(s):  
Jonathan U. Peled ◽  
J. Jessica Yu ◽  
Beibei Belinda Ding ◽  
Rita Shaknovich ◽  
Piotr Sicinski ◽  
...  

Abstract Germinal Centers (GC) of secondary lymphoid tissues are critical to mounting a high-affinity humoral immune response. B cells within the GC undergo rapid clonal expansion and selection while diversifying their antibody genes through class switch recombination and somatic hypermutation. Although it is generally believed that GC B cells employ a unique proliferative program to accommodate these processes, very little is known about how the GC-associated cell cycle is orchestrated. The D-type cyclins are important regulators of the G1 phase of the cell cycle and are the ultimate targets of many mitogenic and oncogenic stimuli. The Cyclin D3 gene is rearranged and over-expressed in certain mature B cell malignancies, and its overexpression has been reported to predict poor clinical outcome in patients with diffuse large B cell lymphoma. It has been observed that during their development, B cells switch from expressing cyclin D2 to cyclin D3 when they are recruited into the GC response. It is unclear, however, whether this switch simply reflects a change in the transcription factors that govern cyclin expression or serves a biological mandate. Here we report that mice deficient in cyclin D3 are profoundly impaired in their ability to form GCs as measured by immunohistochemistry and flow cytometry. Production of antigen-specific antibodies and affinity maturation, as ascertained by ELISA, are concomitantly reduced in these animals. These phenotypes can be at least partially explained by a significant block in the G1-phase of the cell cycle of GC B cells in vivo. Interestingly, this block in the G1-S transition is observed despite an apparent compensatory increase in cyclin D2 expression. In addition, naive B cells activated in vitro by either LPS or LPS and IL-4 display only minor changes in cell-cycle profile, suggesting that a specific requirement for cyclin D3 is unique to GC B cells. We also find moderately reduced Bcl6 mRNA expression in both naïve and GC B cells from the cyclin D3 knockout mice. Since Bcl6 is a master regulator of the GC response, decreased activity of this transcriptional repressor may further contribute to the severity of the GC phenotype. This is the first demonstration that cyclin D3 plays a unique role during the GC response in that it is required for its optimal structure and function. In addition to expanding appreciation for the cell type- and tissue-specific functions of the three D-type cyclin molecules, our findings have implications for understanding the role of Cyclin D3 in human B cell lymphomas.


2008 ◽  
Vol 415 (3) ◽  
pp. 439-448 ◽  
Author(s):  
Katherine A. Kaproth-Joslin ◽  
Xiangquan Li ◽  
Sarah E. Reks ◽  
Grant G. Kelley

In the present study, we examined the role of PLCδ1 (phospholipase C δ1) in the regulation of cellular proliferation. We demonstrate that RNAi (RNA interference)-mediated knockdown of endogenous PLCδ1, but not PLCβ3 or PLCϵ, induces a proliferation defect in Rat-1 and NIH 3T3 fibroblasts. The decreased proliferation was not due to an induction of apoptosis or senescence, but was associated with an approx. 60% inhibition of [3H]thymidine incorporation. Analysis of the cell cycle with BrdU (bromodeoxyuridine)/propidium iodide-labelled FACS (fluorescence-activated cell sorting) demonstrated an accumulation of cells in G0/G1-phase and a corresponding decrease in cells in S-phase. Further examination of the cell cycle after synchronization by serum-starvation demonstrated normal movement through G1-phase but delayed entry into S-phase. Consistent with these findings, G1 cyclin (D2 and D3) and CDK4 (cyclin-dependent kinase 4) levels and associated kinase activity were not affected. However, cyclin E-associated CDK2 activity, responsible for G1-to-S-phase progression, was inhibited. This decreased activity was accompanied by unchanged CDK2 protein levels and paradoxically elevated cyclin E and cyclin E-associated CDK2 levels, suggesting inhibition of the cyclin E–CDK2 complex. This inhibition was not due to altered stimulatory or inhibitory phosphorylation of CDK2. However, p27, a Cip/Kip family CKI (CDK inhibitor)-binding partner, was elevated and showed increased association with CDK2 in PLCδ1-knockdown cells. The result of the present study demonstrate a novel and critical role for PLCδ1 in cell-cycle progression from G1-to-S-phase through regulation of cyclin E–CDK2 activity and p27 levels.


Development ◽  
1993 ◽  
Vol 119 (3) ◽  
pp. 673-690 ◽  
Author(s):  
H.E. Richardson ◽  
L.V. O'Keefe ◽  
S.I. Reed ◽  
R. Saint

We have isolated a Drosophila homolog of the human G1-specific cyclin E gene. Cyclin E proteins thus constitute an evolutionarily conserved subfamily of metazoan cyclins. The Drosophila cyclin E gene, DmcycE, encodes two proteins with a common C-terminal region and unique N-terminal regions. Unlike other Drosophila cyclins, DmcycE exhibits a dynamic pattern of expression during development. DmcycE is supplied maternally, but at the completion of the cleavage divisions and prior to mitosis 14, the maternal transcripts are rapidly degraded in all cells except the pole (germ) cells. Two modes of DmcycE expression are observed in the subsequent divisions. During cycles 14, 15 and 16 in non-neural cells, DmcycE mRNA levels show no cell-cycle-associated variation. DmcycE expression in these cells is therefore independent of the cell cycle phase. In contrast, expression in proliferating embryonic peripheral nervous system cells occurs during interphase as a brief pulse that initiates before and overlaps with S phase, demonstrating the presence of a G1 phase in these embryonic neural cell cycles. DmcycE appears not to be expressed in cells that undergo endoreplication cycles during polytenization. The structural homology to human cyclin E, the ability of DmcycE to rescue a G1 cyclin-deficient yeast strain, the presence of multiple PEST sequences characteristic of G1-specific cyclins and expression during G1 phase in proliferating peripheral nervous system cells all argue that Drosophila cyclin E is a G1 cyclin. Constitutive DmcycE expression in embryonic cycles lacking a G1 phase, in contrast to expression during the G1-S phase transition in cycles exhibiting a G1 phase, implicates DmcycE expression in the regulation of the G1 to S phase transition during Drosophila embryogenesis.


2004 ◽  
Vol 78 (11) ◽  
pp. 5658-5669 ◽  
Author(s):  
Chun-Jen Chen ◽  
Shinji Makino

ABSTRACT Mouse hepatitis virus (MHV) replication in actively growing DBT and 17Cl-1 cells resulted in the inhibition of host cellular DNA synthesis and the accumulation of infected cells in the G0/G1 phase of the cell cycle. UV-irradiated MHV failed to inhibit host cellular DNA synthesis. MHV infection in quiescent 17Cl-1 cells that had been synchronized in the G0 phase by serum deprivation prevented infected cells from entering the S phase after serum stimulation. MHV replication inhibited hyperphosphorylation of the retinoblastoma protein (pRb), the event that is necessary for cell cycle progression through late G1 and into the S phase. While the amounts of the cellular cyclin-dependent kinase (Cdk) inhibitors p21Cip1, p27Kip1, and p16INK4a did not change in infected cells, MHV infection in asynchronous cultures induced a clear reduction in the amounts of Cdk4 and G1 cyclins (cyclins D1, D2, D3, and E) in both DBT and 17Cl-1 cells and a reduction in Cdk6 levels in 17Cl-1 cells. Infection also resulted in a decrease in Cdk2 activity in both cell lines. MHV infection in quiescent 17Cl-1 cells prevented normal increases in Cdk4, Cdk6, cyclin D1, and cyclin D3 levels after serum stimulation. The amounts of cyclin D2 and cyclin E were not increased significantly after serum stimulation in mock-infected cells, whereas they were decreased in MHV-infected cells, suggesting the possibility that MHV infection may induce cyclin D2 and cyclin E degradation. Our data suggested that a reduction in the amounts of G1 cyclin-Cdk complexes in MHV-infected cells led to a reduction in Cdk activities and insufficient hyperphosphorylation of pRb, resulting in inhibition of the cell cycle in the G0/G1 phase.


2005 ◽  
Vol 25 (21) ◽  
pp. 9292-9303 ◽  
Author(s):  
Taichi Hara ◽  
Takumi Kamura ◽  
Shuhei Kotoshiba ◽  
Hidehisa Takahashi ◽  
Kenichiro Fujiwara ◽  
...  

ABSTRACT KPC2 (Kip1 ubiquitylation-promoting complex 2) together with KPC1 forms the ubiquitin ligase KPC, which regulates degradation of the cyclin-dependent kinase inhibitor p27 at the G1 phase of the cell cycle. KPC2 contains a ubiquitin-like (UBL) domain, two ubiquitin-associated (UBA) domains, and a heat shock chaperonin-binding (STI1) domain. We now show that KPC2 interacts with KPC1 through its UBL domain, with the 26S proteasome through its UBL and NH2-terminal UBA domains, and with polyubiquitylated proteins through its UBA domains. The association of KPC2 with KPC1 was found to stabilize KPC1 in a manner dependent on the STI1 domain of KPC2. KPC2 mutants that lacked either the NH2-terminal or the COOH-terminal UBA domain supported the polyubiquitylation of p27 in vitro, whereas a KPC2 derivative lacking the STI1 domain was greatly impaired in this regard. Depletion of KPC2 by RNA interference resulted in inhibition of p27 degradation at the G1 phase, and introduction of KPC2 derivatives into the KPC2-depleted cells revealed that the NH2-terminal UBA domain of KPC2 is essential for p27 degradation. These observations suggest that KPC2 cooperatively regulates p27 degradation with KPC1 and that the STI1 domain as well as the UBL and UBA domains of KPC2 are indispensable for its function.


Development ◽  
1998 ◽  
Vol 125 (24) ◽  
pp. 5069-5078 ◽  
Author(s):  
J. Horsfield ◽  
A. Penton ◽  
J. Secombe ◽  
F.M. Hoffman ◽  
H. Richardson

During eye development in Drosophila, cell cycle progression is coordinated with differentiation. Prior to differentiation, cells arrest in G1 phase anterior to and within the morphogenetic furrow. We show that Decapentaplegic (Dpp), a TGF-β family member, is required to establish this G1 arrest, since Dpp-unresponsive cells located in the anterior half of the morphogenetic furrow show ectopic S phases and ectopic expression of the cell cycle regulators Cyclins A, E and B. Conversely, ubiquitous over-expression of Dpp in the eye imaginal disc transiently inhibits S phase without affecting Cyclin E or Cyclin A abundance. This Dpp-mediated inhibition of S phase occurs independently of the Cyclin A inhibitor Roughex and of the expression of Dacapo, a Cyclin E-Cdk2 inhibitor. Furthermore, Dpp-signaling genes interact genetically with a hypomorphic cyclin E allele. Taken together our results suggest that Dpp acts to induce G1 arrest in the anterior part of the morphogenetic furrow by a novel inhibitory mechanism. In addition, our results provide evidence for a Dpp-independent mechanism that acts in the posterior part of the morphogenetic furrow to maintain G1 arrest.


1998 ◽  
Vol 18 (12) ◽  
pp. 7584-7589 ◽  
Author(s):  
Kwang-Ai Won ◽  
Robert J. Schumacher ◽  
George W. Farr ◽  
Arthur L. Horwich ◽  
Steven I. Reed

ABSTRACT Cyclin E, a partner of the cyclin-dependent kinase Cdk2, has been implicated in positive control of the G1/S phase transition. Whereas degradation of cyclin E has been shown to be exquisitely regulated by ubiquitination and proteasomal action, little is known about posttranscriptional aspects of its biogenesis. In a yeast-based screen designed to identify human proteins that interact with human cyclin E, we identified components of the eukaryotic cytosolic chaperonin CCT. We found that the endogenous CCT complex in yeast was essential for the maturation of cyclin E in vivo. Under conditions of impaired CCT function, cyclin E failed to accumulate. Furthermore, newly translated cyclin E, both in vitro in reticulocyte lysate and in vivo in human cells in culture, is efficiently bound and processed by the CCT. In vitro, in the presence of ATP, the bound protein is folded and released in order to become associated with Cdk2. Thus, both the acquisition of the native state and turnover of cyclin E involve ATP-dependent processes mediated by large oligomeric assemblies.


Sign in / Sign up

Export Citation Format

Share Document