scholarly journals Headless flies generated by developmental pathway interference

Development ◽  
2001 ◽  
Vol 128 (17) ◽  
pp. 3307-3319 ◽  
Author(s):  
Renjie Jiao ◽  
Michael Daube ◽  
Hong Duan ◽  
Yu Zou ◽  
Erich Frei ◽  
...  

Ectopic expression of transcription factors in eye-antennal discs of Drosophila strongly interferes with their developmental program. Early ectopic expression in embryonic discs interferes with the developmental pathway primed by Eyeless and generates headless flies, which suggests that Eyeless is necessary for initiating cell proliferation and development of both the eye and antennal disc. Interference occurs through a block in the cell cycle that for some ectopic transcription factors is overcome by D-CycE or D-Myc. Late ectopic expression in cone cell precursors interferes with their differentiation. We propose that this developmental pathway interference is a general surveillance mechanism that eliminates most aberrations in the genetic program during development and evolution, and thus seriously restricts the pathways that evolution may take.

2021 ◽  
Vol 20 ◽  
Author(s):  
Rabih Roufayel ◽  
Rabih Mezher ◽  
Kenneth B. Storey

: Selected transcription factors have critical roles to play in organism survival by regulating the expression of genes that control the adaptations needed to handle stress conditions. The retinoblastoma (Rb) protein coupled with the E2F transcription factor family was demonstrated to have roles in controlling the cell cycle during freezing and associated environmental stresses (anoxia, dehydration). Rb phosphorylation or acetylation at different sites provide a mechanism for repressing cell proliferation that is under the control of E2F transcription factors in animals facing stresses that disrupt cellular energetics or cell volume controls. Other central regulators of the cell cycle including Cyclins, Cyclin dependent kinases (Cdks), and checkpoint proteins detect DNA damage or any improper replication, blocking further progression of cell cycle and interrupting cell proliferation. This review provides an insight into the molecular regulatory mechanisms of cell cycle control, focusing on Rb-E2F along with Cyclin-Cdk complexes typically involved in development and differentiation that need to be regulated in order to survive extreme cellular stress.


PPAR Research ◽  
2007 ◽  
Vol 2007 ◽  
pp. 1-10 ◽  
Author(s):  
A. Cimini ◽  
L. Cristiano ◽  
E. Benedetti ◽  
B. D'Angelo ◽  
M. P. Cerù

PPAR isotypes are involved in the regulation of cell proliferation, death, and differentiation, with different roles and mechanisms depending on the specific isotype and ligand and on the differentiated, undifferentiated, or transformed status of the cell. Differentiation stimuli are integrated by key transcription factors which regulate specific sets of specialized genes to allow proliferative cells to exit the cell cycle and acquire specialized functions. The main differentiation programs known to be controlled by PPARs both during development and in the adult are placental differentiation, adipogenesis, osteoblast differentiation, skin differentiation, and gut differentiation. PPARs may also be involved in the differentiation of macrophages, brain, and breast. However, their functions in this cell type and organs still awaits further elucidation. PPARs may be involved in cell proliferation and differentiation processes of neural stem cells (NSC). To this aim, in this work the expression of the three PPAR isotypes and RXRs in NSC has been investigated.


2002 ◽  
Vol 22 (22) ◽  
pp. 7842-7852 ◽  
Author(s):  
Marc Schmidt ◽  
Sylvia Fernandez de Mattos ◽  
Armando van der Horst ◽  
Rob Klompmaker ◽  
Geert J. P. L Kops ◽  
...  

ABSTRACT The FoxO forkhead transcription factors FoxO4 (AFX), FoxO3a (FKHR.L1), and FoxO1a (FKHR) represent important physiological targets of phosphatidylinositol-3 kinase (PI3K)/protein kinase B (PKB) signaling. Overexpression or conditional activation of FoxO factors is able to antagonize many responses to constitutive PI3K/PKB activation including its effect on cellular proliferation. It was previously shown that the FoxO-induced cell cycle arrest is partially mediated by enhanced transcription and protein expression of the cyclin-dependent kinase inhibitor p27kip1 (R. H. Medema, G. J. Kops, J. L. Bos, and B. M. Burgering, Nature 404:782-787, 2000). Here we have identified a p27kip1-independent mechanism that plays an important role in the antiproliferative effect of FoxO factors. Forced expression or conditional activation of FoxO factors leads to reduced protein expression of the D-type cyclins D1 and D2 and is associated with an impaired capacity of CDK4 to phosphorylate and inactivate the S-phase repressor pRb. Downregulation of D-type cyclins involves a transcriptional repression mechanism and does not require p27kip1 function. Ectopic expression of cyclin D1 can partially overcome FoxO factor-induced cell cycle arrest, demonstrating that downregulation of D-type cyclins represents a physiologically relevant mechanism of FoxO-induced cell cycle inhibition.


2021 ◽  
Author(s):  
Stefan S Thor ◽  
Behzad Yaghmaeian Salmani ◽  
Brad Balderson ◽  
Susanne Bauer ◽  
Helen Ekman ◽  
...  

The hypothalamus displays staggering cellular diversity, chiefly established during embryogenesis by the interplay of several signalling pathways and a battery of transcription factors. However, the contribution of epigenetic cues to hypothalamus development remains unclear. We mutated the Polycomb Repressor Complex 2 gene Eed in the developing mouse hypothalamus, which resulted in the loss of H3K27me3; a fundamental epigenetic repressor mark. This triggered ectopic expression of posteriorly expressed regulators (e.g., Hox homeotic genes), upregulation of cell cycle inhibitors and reduced proliferation. Surprisingly, despite these effects, single cell transcriptomic analysis revealed that the majority of neuronal subtypes were still generated in Eed mutants. However, we observed an increase in Glutamatergic/GABAergic double-positive cells, as well as loss/reduction of dopamine, Hypocretin/Orexin and Tac2 neurons. These findings indicate that many aspects of the hypothalamic gene regulatory flow can proceed without the key H3K27me3 epigenetic repressor mark, and points to a unique sensitivity of particular neuronal sub-types to a disrupted epigenomic landscape.


Nutrients ◽  
2020 ◽  
Vol 12 (8) ◽  
pp. 2480
Author(s):  
Phutthida Kongthitilerd ◽  
Tanyawan Suantawee ◽  
Henrique Cheng ◽  
Thavaree Thilavech ◽  
Marisa Marnpae ◽  
...  

Riceberry rice (Oryza sativa L.) is a new pigmented variety of rice from Thailand. Despite its high anthocyanin content, its effect on adipogenesis and adipocyte function remains unexplored. We investigated whether Riceberry rice extract (RBE) impacted cell proliferation by examining viability and cell cycle, using preadipocyte 3T3-L1 cells. To test RBE’s effect on adipocyte formation, cells were cultured in adipogenic medium supplemented with extract and adipocyte number and triglyceride levels were quantified. Furthermore, Akt1 phosphorylation along with RT-qPCR and intracellular calcium imaging were performed to obtain an insight into its mechanism of action. The effect of RBE on adipocyte function was investigated using glucose uptake and lipolysis assays. Treatment of cells with RBE decreased preadipocyte number without cytotoxicity despite inducing cell cycle arrest (p < 0.05). During adipogenic differentiation, RBE supplementation reduced adipocyte number and triglyceride accumulation by downregulating transcription factors (e.g., PPARγ, C/EBPα, and C/EBPβ) and their target genes (p < 0.05). The Akt1 phosphorylation was decreased by RBE but insignificance, however, the extract failed to increase intracellular calcium signals. Finally, the treatment of adipocytes with RBE reduced glucose uptake by downregulating Glut4 mRNA expression and enhanced isoproterenol-induced lipolysis (p < 0.05). These findings suggest that RBE could potentially be used in the treatment of obesity by inhibiting adipocyte formation and proliferation.


2012 ◽  
Vol 108 (07) ◽  
pp. 148-158 ◽  
Author(s):  
Shailaja Mahajan ◽  
Anke Fender ◽  
Jutta Meyer-Kirchrath ◽  
Muhammed Kurt ◽  
Mareike Barth ◽  
...  

SummaryThrombin exerts coagulation-independent effects on the proliferation and migration of vascular smooth muscle cells (SMC). Forkhead box-O (FoxO) transcription factors regulate cell proliferation, apoptosis and cell cycle arrest, but a possible functional interaction between thrombin and FoxO factors has not been identified to date. In human cultured vascular SMC, thrombin induced a time-dependent phosphorylation of FoxO1 and FoxO3 but not FoxO4. This effect was mimicked by an activating-peptide (AP) for protease-activated receptor (PAR)-1, and abolished by a PAR-1 antagonist (SCH79797). APs for other PARs were without effect. FoxO1 and FoxO3 phosphorylation were prevented by the PI3 kinase (PI3K) inhibitor LY294002 while inhibitors of ERK1/2 (PD98059) or p38MAPK (SB203580) were ineffective. LY294002 moreover prevented thrombin-stimulated SMC mitogenesis and proliferation. FoxO1 and FoxO3 siRNA augmented basal DNA synthesis and proliferation of SMC. Nuclear content of FoxO proteins decreased time-dependently in response to thrombin, coincided with suppressed expression of the cell cycle regulating genes p21CIP1 and p27kip1 by thrombin. FoxO1 siRNA reduced basal p21CIP1 while FoxO3 siRNA attenuated p27kip1 expression; thrombin did not show additive effects. LY294002 restored p21CIP1 and p27kip1 protein expression. Immunohistochemistry revealed that human native and failed saphenous vein grafts were characterised by the cytosolic presence of p-FoxO factors in co-localisation of p21CIP1 and p27kip1 with SMC. In conclusion, thrombin and FoxO factors functionally interact through PI3K/Akt-dependent FoxO phosphorylation leading to expression of cell cycle regulating genes and ultimately SMC proliferation. This may contribute to remodelling and failure of saphenous vein bypass grafts.


2014 ◽  
Vol 96 (3) ◽  
pp. 453-462 ◽  
Author(s):  
Johannes Wedel ◽  
Maximillia C. Hottenrott ◽  
Eleni Stamellou ◽  
Annette Breedijk ◽  
Charalambos Tsagogiorgas ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3680-3680
Author(s):  
Ujunwa Cynthia Okoye-Okafor ◽  
Laura Barreyro ◽  
Heng Rui Wang ◽  
Boris Bartholdy ◽  
Britta Will ◽  
...  

Abstract Cell cycling is a tightly regulated process involving the structured expression modulation of various regulatory genes. This process is crucial for the maintenance of cell survival/proliferation in both normal and malignant hematopoietic cells. We have previously described the highly expressed CIITA-BX648577 gene fusion (Steidl C. et al., Nature 2011), involving the novel gene locus BX648577/FLJ27352 /hypothetical LOC 145788/C15orf65 and the Class II Transactivator (CIITA) in the Hodgkin’s lymphoma cell line KM-H2. While CIITA is well known to be involved in the regulation of immune responses, specifically through regulation of the Major Histocompatibility Complex (MHC)-II, nothing is known about the expression and function of the BX648577 locus. The objective of the current study was to (I) study RNA and protein expression of the putative full length gene encoded by the BX648577 (TIHL) gene locus, and (II) study its biological function in normal and malignant hematopoietic cells, including its effects on cell proliferation, clonogenicity and cell death. We detected robust endogenous TIHL RNA and protein expression in a variety of healthy and malignant hematopoietic cell types using quantitative real-time polymerase chain reaction (qRT-PCR) and western blot analysis using a TIHL-specific antibody. At the functional level, we found that ectopic expression of the highly conserved full length TIHL protein in human NB4 leukemia cells and murine hematopoietic progenitor HPC-7 cells leads to enhanced clonogenicity and increased proliferative capacity with significant increases in the percentage of cells in S-phase of the cell cycle. Furthermore, we observed more aggressive leukemia and decreased survival of NSG mice following retro-orbital transplantation of TIHL-expressing compared to empty control-expressing NB4 cells. Interestingly, although we did not observe a change in the rate of cell proliferation or colony forming ability following TIHL overexpression in the ATRA-resistant cell line NB4.306, there was a significant alteration in its cell cycle distribution, with an increase in the fraction of cells in S-phase. The increase in S-phase cells was confirmed by 5-ethynyl-2’-deoxyuridine (EdU) incorporation assays and flow cytometry. Similarly, knockdown of TIHL using 2 independent lentiviral shRNAs, led to a significant decrease in the growth of both NB4 and acute myeloid leukemia KG1a cells in both suspension cultures and semi-solid media. Although we observed slightly increased apoptosis upon TIHL downregulation, the changes could be more significantly attributed to a decrease in the percentage of cells in S-phase within 2-3 days after transduction with the lentiviral shRNAs. Finally, in silico analysis of the TIHL promoter identified various predicted transcriptional regulators of TIHL, the majority of which are cell cycle specific transcription factors including Nuclear Receptor Subfamily 5 Group A Member 1 (NR5A1), the ets domain transcription factors ELF5 and ELF1, and Glioma-Associated Oncogene Homolog 1 (GLI-1). Our findings thus far strongly support a novel role for TIHL in cell cycle regulation/modulation in both normal and malignant hematopoiesis. Future directions include gene expression studies to identify downstream targets of TIHL following overexpression and knockdown and co-immunoprecipitation coupled to mass spectrometry analysis will be used to identify direct interacting protein partners of this novel gene. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 20 (1) ◽  
Author(s):  
Chang Yang ◽  
Zhao-Cong Zhang ◽  
Tian -Bo Liu ◽  
Ye Xu ◽  
Bai-Rong Xia ◽  
...  

Abstract Background Cervical cancer is the second leading cause of death in women 20–39 years old. Because coverage for cervical cancer screening is low, and the vaccination rate of human papillomavirus (HPV) is poor in some countries, potential markers to detect the disease at early stages are needed. E2F transcription factors (E2Fs) are a family of transcription factors that function in cell proliferation, differentiation, apoptosis, and tumorigenesis. As abnormal activation and regulation of E2Fs are related to tumor development and poor prognosis, we performed bioinformatic analyses and in vitro assays to evaluate the role of E2Fs in cervical cancer. Methods Transcriptional expression of E2Fs was initially evaluated in silico using ONCOMINE and Gene Expression Profiling Interactive Analysis (GEPIA), followed by evaluation of E2F1/2/7/8 protein levels using immunohistochemistry in 88 patient tissues. E2F2 and E2F7 mRNA levels were measured by RT-qPCR. LinkedOmics and Metascape were used to predict functions of E2Fs, and in vitro experiments were performed to assess the tumorigenic role of E2F2 and E2F7. Results In silico analysis showed that E2F1/2/7/8 were significantly overexpressed in cervical cancer, findings which were confirmed at the protein level using immunohistochemistry. Further, upregulation of E2F1/2/7/8 was associated with different clinicopathological prognostic factors, including positivity for lymph vessel invasion and deep invasion of cervical stroma. Increased expression of E2F1/2/7/8 was also related to shorter overall survival (OS) and disease-free survival (DFS) in patients with cervical cancer. Using multivariate analysis, we confirmed E2F1/2/7/8 as independent prognostic factors for shorter OS of patients with cervical cancer. Finally, in vitro experiments showed that E2F2 and E2F7 are involved in cell proliferation and migration and cell cycle regulation in both HPV-positive and HPV-negative cervical cancer cells. Conclusions E2F1/2/7/8 may be prognostic biomarkers for survival of patients with cervical cancer. E2F2 and E2F7 are involved in cell proliferation, migration, and cell cycle in both HPV-positive and HPV-negative cervical cancer cells.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Margaret Yeh ◽  
Yin-Ying Wang ◽  
Ji Young Yoo ◽  
Christina Oh ◽  
Yoshihiro Otani ◽  
...  

AbstractTumor suppressive microRNAs (miRNAs) are increasingly implicated in the development of anti-tumor therapy by reprogramming gene network that are aberrantly regulated in cancer cells. This study aimed to determine the therapeutic potential of putative tumor suppressive miRNA, miR-138, against glioblastoma (GBM). Whole transcriptome and miRNA expression profiling analyses on human GBM patient tissues identified miR-138 as one of the significantly downregulated miRNAs with an inverse correlation with CD44 expression. Transient overexpression of miR-138 in GBM cells inhibited cell proliferation, cell cycle, migration, and wound healing capability. We unveiled that miR-138 negatively regulates the expression of CD44 by directly binding to the 3′ UTR of CD44. CD44 inhibition by miR-138 resulted in an inhibition of glioblastoma cell proliferation in vitro through cell cycle arrest as evidenced by a significant induction of p27 and its translocation into nucleus. Ectopic expression of miR-138 also increased survival rates in mice that had an intracranial xenograft tumor derived from human patient-derived primary GBM cells. In conclusion, we demonstrated a therapeutic potential of tumor suppressive miR-138 through direct downregulation of CD44 for the treatment of primary GBM.


Sign in / Sign up

Export Citation Format

Share Document