scholarly journals Inhibition of Aurora Kinase B activity disrupts development and differentiation of salivary glands

2021 ◽  
Vol 7 (1) ◽  
Author(s):  
Abeer K. Shaalan ◽  
Tathyane H. N. Teshima ◽  
Abigail S. Tucker ◽  
Gordon B. Proctor

AbstractLittle is known about the key molecules that regulate cell division during organogenesis. Here we determine the role of the cell cycle promoter aurora kinase B (AURKB) during development, using embryonic salivary glands (E-SGs) as a model. AURKB is a serine/threonine kinase that regulates key events in mitosis, which makes it an attractive target for tailored anticancer therapy. Many reports have elaborated on the role of AURKB in neoplasia and cancer; however, no previous study has shown its role during organ development. Our previous experiments have highlighted the essential requirement for AURKB during adult exocrine regeneration. To investigate if AURKB is similarly required for progression during embryonic development, we pharmacologically inhibited AURKB in developing submandibular glands (SMGs) at embryonic day (E)13.5 and E16.5, using the highly potent and selective drug Barasertib. Inhibition of AURKB interfered with the expansion of the embryonic buds. Interestingly, this effect on SMG development was also seen when the mature explants (E16.5) were incubated for 24 h with another cell cycle inhibitor Aphidicolin. Barasertib prompted apoptosis, DNA damage and senescence, the markers of which (cleaved caspase 3, γH2AX, SA-βgal and p21, respectively), were predominantly seen in the developing buds. In addition to a reduction in cell cycling and proliferation of the epithelial cells in response to AURKB inhibition, Barasertib treatment led to an excessive generation of reactive oxygen species (ROS) that resulted in downregulation of the acinar differentiation marker Mist1. Importantly, inhibition of ROS was able to rescue this loss of identity, with Mist1 expression maintained despite loss of AURKB. Together, these data identify AURKB as a key molecule in supporting embryonic development and differentiation, while inhibiting senescence-inducing signals during organogenesis.

EMJ Oncology ◽  
2021 ◽  
pp. 81-90
Author(s):  
Salini Das ◽  
Elizabeth Mahapatra ◽  
Souvick Biswas ◽  
Madhumita Roy ◽  
Sutapa Mukherjee

Radiotherapy is one of the most conventional modes of treatment in several cancers. Failure of radiotherapy followed by acquisition of radioresistance is one of the emerging challenges faced by clinical experts. Unusual expression and functional implications of several molecules are observed to facilitate radioresistance. Aurora A, a member of the Aurora kinase (serine/threonine kinase) family, is one such molecule that shows significantly altered expression as well as non-canonical functional crosstalk with other associated factors (cell cycle regulators, signaling molecules, stemness markers, etc.) to favour the adaptations for the acquirement of radioresistance. These mechanisms include progression of cell cycle, stimulatory activation of factors by phosphorylation for enhancing the chance of cellular survivability, and prevention of apoptosis. This review article summarises how Aurora A is responsible for radioresistance in cancer and why this kinase should be considered a negative biomarker of radiosensitivity. This review discloses a wider opportunity in the field of research to find the mechanistic key regulatory pathway of Aurora A, which can be a potential target for enhancing the efficiency of treatment. Further investigations are required to explore the potential of Aurora A inhibitors as reliable radiosensitisers.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 888-888 ◽  
Author(s):  
Okinaka Keiji ◽  
Satoki Nakamura ◽  
Isao Hirano ◽  
Takaaki Ono ◽  
Shinya Fujisawa ◽  
...  

Abstract [Background] FoxM1, a member of the Fox transcription factor family, plays an important cell cycle regulator of both the transition from G1 to S phase and progression to mitosis. FoxM1 expression was also found to be up-regulated in some solid tumors (basal cell carcinomas, hepatocellular carcinoma, and primary breast cancer). These results suggested that FoxM1 plays a role in the oncogenesis of malignancies. However, it is unknown whether FoxM1 expression contributes to the development or progression of leukemia cells. Therefore, we investigated how FoxM1 regulated the cell cycle of leukemia cells and the expression analysis of the FoxM1 gene in patients with acute leukemias. [Methods] The cells used in this study were human acute leukemia cell lines, U937 and YRK2 cells. Primary acute myeloblastic (25 AML (4 M1, 11 M2, 6 M4, 4 M5)) cells were obtained from the peripheral blood. Human normal mononuclear cells (MNCs) were isolated from peripheral blood (PB) of healthy volunteers after obtaining informed consents. For analysis of proliferation and mitotic regulatory proteins (p27, p21, Skp2, Cdc25B, Cyclin D1, Survivin, Aurora kinase B, and KIS) in leukemia cells, MTT assays and western blot were performed in all cell lines, which untransfected or transfected with siRNA FoxM1, respectively. For cell cycle analysis, flow cytometory analysis was performed in leukemia cells untransfected or transfected with siRNAFoxM1 by PI staining. For analysis of FoxM1 mRNA, quantitative RT-PCR was performed in all cell lines and clinical samples. [Results] In all leukemia cell lines, the expression of FoxM1B mRNA were significantly higher than normal MNCs. When transfected with the siRNA FoxM1 in leukemia cells, suppression of FoxM1 caused a mean 71% (range 62 to 80%) reduction in S phase cells and a mean 4.4-fold (range 3.2 to 5.6-fold) increase in G2/M phase cells compared to controls. MTT assay demonstrated that the proliferation of the siRNA FoxM1 transfected cells was inhibited compared to the untransfected cells. Moreover, FoxM1 knockdown by siRNA in leukemia cells reduced protein and mRNA expression of Aurora kinase B, Survivin, Cyclin D1, Skp2 and Cdc25B, while increased protein expression of p21and p27. In the clinical samples obtained from patients with acute leukemias, the FoxM1B gene was overexpressed in 22/25 (88%). The relative folds of FoxM1B gene expression were for AML: 2.83 compared to normal MNCs. [Conclusions] In this study, we report in the first time that FoxM1 is overexpressed in myeloid leukemia cells. These results demonstrated that expression of FoxM1 is an essential transcription factor for growth of leukemia cells, and regulate expression of the mitotic regulators. Moreover, we showed that FoxM1 induced the expression of KIS protein. Therefore, FoxM1 might be one of moleculer targets of therapy for acute leukemias.


Development ◽  
1988 ◽  
Vol 104 (3) ◽  
pp. 415-422
Author(s):  
C. Aimar

The role of the cytoplasm and nucleus in the control of the length of the division cycle was investigated in Pleurodeles waltl eggs. Injection of spermine into enucleated eggs showed that the ability to cleave was not restricted just to the period of normal cytokinesis (T=1.0) but was possible throughout most of the first egg cycle. The cytoplasmic components required for cytokinesis seem to increase progressively during the first division cycle. Nuclear transfer experiments indicated that the timing of cleavage was normal only when the nucleus and egg cytoplasm were reassociated between T=0.0 (activation time) and T=0.50. Delayed associations, after T=0.50, provoked an alteration in the chronology of first cleavage and led to abnormal embryonic development. In the absence of a nucleus, the egg cycle seemed to stop at T=0.50. These different observations suggest that the normal timing of cleavage not only depends on a ‘cytoplasmic clock’ but is also determined by an isochronous nucleocytoplasmic relationship during the early phase of egg development.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4320-4320
Author(s):  
Satoki Nakamura ◽  
Takaaki Ono ◽  
Yuya Sugimoto ◽  
Miki Kobayashi ◽  
Naohi Sahara ◽  
...  

Abstract [Background] FoxM1, a member of the Fox transcription factor family, plays an important cell cycle regulator of both the transition from G1 to S phase and progression to mitosis. FoxM1 expression was also found to be up-regulated in some solid tumors (basal cell carcinomas, hepatocellular carcinoma, and primary breast cancer). These results suggested that FoxM1 plays a role in the oncogenesis of malignancies. However, it is unknown whether FoxM1 expression contributes to the development or progression of leukemia cells. Therefore, we investigated whether and how FoxM1 regulated the cell cycle of leukemia cells. [Methods] The cells used in this study were human leukemia cell lines, K562, HL60, U937 cells. For analysis of FoxM1 mRNA, RT-PCR was performed in all cell lines. For analysis of proliferation and mitotic regulatory proteins (p27, p21, Skp2, Cdc25B, Cyclin D1, Survivin, and Aurora kinase B) in leukemia cells, MTT assays and western blot were performed in all cell lines untransfected or transfected with siRNA FoxM1, respectively. For cell cycle analysis, flow cytometory analysis was performed in leukemia cells untransfected or transfected with siRNAFoxM1 by PI staining. [Results] In all leukemia cell lines, the expression of FoxM1B mRNA were significantly higher than normal MNCs. In K562, HL60, and U937 cells transfected with the siRNA FoxM1, suppression of FoxM1 caused a mean 71% (range 62 to 80%) reduction in S phase cells and a mean 4.4-fold (range 3.2 to 5.6-fold) increase in G2/M phase cells compared to untransfected cells. MTT assay demonstrated that the proliferation of the siRNA FoxM1 transfected cells was inhibited compared to the untransfected cells at 2, 3, 4, or 5 days after siRNA FoxM1 transfection. FoxM1 has been reported to regulate transcription of essential mitotic regulatory genes. We showed that FoxM1 knockdown by siRNA in leukemia cells reduced protein and mRNA expression of Aurora kinase B, Survivin, Cyclin D1, Skp2 and Cdc25B, while increased protein expression of p21and p27 in RT-PCR and western blot analysis. [Conclusions] In this study, we report in the first time that FoxM1 is overexpressed in myeloid leukemia cells. These results demonstrated that expression of FoxM1 is an essential transcription factor for growth of leukemia cells, and regulate expression of the mitotic regulators, Cdc25B, Cyclin D1, Survivin, Aurora kinase B, and p21. Moreover, we showed that FoxM1 regulated the expression of Skp2 protein, which is known to promote degradation of the cell cycle regulator p27. Our study found that inhibition of FoxM1 expression in leukemia cells suppressed their growth in vitro. Therefore, FoxM1 might be a new potential target of therapy for leukemias. We will have further study whether the level of FoxM1 expression in leukemia cells is correlated with patient survival or sensitivity for chemotherapy.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1690-1690
Author(s):  
Leo Kretzner ◽  
Anna Scuto ◽  
Kowolik Claudia ◽  
Richard Jove ◽  
Stephen J Forman ◽  
...  

Abstract Abstract 1690 Poster Board I-716 Background Patients with relapsed or refractory Hodgkin (HL) and Non Hodgkin Lymphoma (NHL) have few options after salvage therapy and transplant, and new agents are thus needed. MK-5108 is a novel aurora kinase inhibitor (AKI) with specificity against aurora kinase A, that produces G2/M phase cell cycle arrest. We show that addition of vorinostat, a histone and protein deacetylase inhibitor, to AKI treatment results in reactivation of proapoptotic genes and enhanced lymphoma cell death. A panel of HL and NHL cell lines was studied with either drug or the combination, using cell growth, apoptosis, and flow cytometry assays, followed by molecular studies. Results MK-5108 alone at 0.1 – 3 mM results in significant growth inhibition and apoptosis in multiple cell lines representing Hodgkin, Burkitt, and Non-Hodgkin lymphoma types, interestingly,DHL-4 and DHL-6 cells were more sensitive to this agent than to the pan-AKI MK-0457. Vorinostat alone at a dose range of 0.5 – 3 mM reduces cell growth by 50% or more in all lines tested. The combination of 1.5 mM vorinostat and 100 nM MK-5108 results in over 85% apoptosis of multiple lymphoma lines tested at 72 hours. Cell cycle analyses by FACS of MK-5108 treated cells show an increased percentage of cells in G2/M with few cells in sub-G1, whereas in combination with vorinostat the G2/M peak decreases and there is a significant increase in the apoptotic sub-G1 population. Real-time PCR analysis and immunoblotting of L540 cells treated with either single agent or in combination revealed that vorinostat treatment leads to alteration in pro-apoptosis, growth arrest, and DNA damage response genes. Myc mRNA and protein levels are reduced by vorinostat, and repression of microRNAs (miRNAs) in the Myc-regulated polycistronic cluster of miRNAs of chromosome 13, such as miR-17.5p, -17.3p, and 18, occurs with vorinostat and TSA. Prosurvival genes such as bcl-XL and hTERT are downregulated five-fold by vorinostat treatment, while the proapoptotic BAK gene is upregulated 1.5 – 2-fold. Vorinostat treatment leads to enhanced acetylation of p53, with a corresponding increase in the p53 target genes p21 and Noxa. To analyze the role of Myc inhibition in the sensitization by vorinostat of lymphoma cells to MK-5108, siRNA-mediated knock-down of Myc expression in L540 cells was performed. The siRNA-Myc transfected L540 cells showed enhanced sensitivity to MK-5108 as compared to control siRNA-null cells, as well as decreased hTERT levels, confirming the role of Myc inhibition by vorinostat as an integral part of the sensitization of lymphoma cells to MK-5108. Conclusions The HDACi vorinostat leads to both transcriptional and post-transcriptional changes that create a pro-apoptotic milieu, sensitizing the cell to centrosome-acting agents such as the aurora kinase A inhibitor MK-5108. These preclinical data support clinical trials of MK-5108 plus vorinostat in patients with relapsed or refractory lymphomas. [We acknowledge Merck Inc for providing Vorinostat, MK-0457, MK-5108, and research support.] Disclosures Kretzner: Merck: Research Funding. Yen:Merck: Research Funding. Kirschbaum:Merck: Research Funding, Speakers Bureau.


2020 ◽  
Vol 22 (Supplement_3) ◽  
pp. iii302-iii303
Author(s):  
Hannah Chatwin ◽  
Rakeb Lemma ◽  
John DeSisto ◽  
Aaron Knox ◽  
Shelby Mestnik ◽  
...  

Abstract Diffuse intrinsic pontine glioma (DIPG) is a fatal pediatric brain tumor with no curative treatments. Approximately 80% of DIPGs contain an H3K27M mutation. The implications of the mutation and how they may be targeted are not fully understood. We established an H3K27M effect-isolating model by transducing H3K27-wildtype lines (HSJD-GBM-001, normal human astrocytes) with lentiviral-packaged H3K27M. We characterized H3K27M-related changes through western blot, phenotypic assays, and RNA-seq. Drug screening of H3K27-wildtype and matched H3K27M-transduced lines was used to identify targets more effective with H3K27M present. Patient-derived pediatric glioblastoma and DIPG lines (BT-245, SU-DIPG-IV, HSJD-DIPG-007, SU-DIPG-XIII*, SF7761) were used for validation. We observed increased H3K27ac and decreased H3K27me3, as well as increased proliferative and migratory abilities, with the addition of H3K27M to H3K27-wildtype lines. RNA-seq showed downregulation of cell cycle regulation and upregulation of epithelial-mesenchymal transition. GSK1070916, an Aurora kinase B/C inhibitor, was isolated from a synthetic lethality screen with H3K27M. GSK1070916 showed strong efficacy in native H3K27M lines (IC50s=60nM-1250nM), superior to the Aurora kinase A inhibitor alisertib, to which all cell lines showed substantial resistance. Combination of both drugs was not synergistic. GSK1070916 treatment caused increased H3K27me3 and decreased H3S10ph and H3S28ph. GSK1070916 induced apoptosis and S-phase stall. The H3K27M mutation induces epigenetic, phenotypic, and cell cycle regulation changes resulting in relaxation of transcriptional controls and more aggressive growth. Aurora kinase B/C inhibition is a novel therapeutic modality for DIPG that appears capable of reversing some H3K27M-related epigenetic changes, inducing apoptosis, and repressing uncontrolled cellular division.


2017 ◽  
Vol 4 (4) ◽  
pp. 1380 ◽  
Author(s):  
Jiahai Chen ◽  
Xiaoli Yang

Background: Hepatocellular carcinoma (HCC) is one of the most frequent malignant tumors. The objective was to investigate the role of serine/threonine kinase Pim-2 in apoptosis signal transduction pathway, because there is little study about its contribution to apoptosis in hepatocellular carcinoma.Methods: The Pim-2 gene and protein expression were examined by qRT-PCR, Western blot and immunohistochemical stain in HCC tissues and normal liver tissues. The plasmid pCI-neo-Pim2 was transfected into human hepatoma cell line SMMC7721 by lipofectamine. Total RNAs were extracted from SMMC7721 cell in logarithm growth phase. The mRNA expression of Pim-2, Akt-1 (protein kinase B), 4E-BP1 (translation repressor of mammalian target of rapamycln), SOCS-1 (repressor of cytokine), Bad(Bcl-xL/Bcl-2 associated death promoter, Bim(Bc1-2 interacting mediator of cell death)and Puma (p53 upregulated modulator of apoptosis) were identified by qRT-PCR. The cell cycle of post-transfected SMMC7721 cells was assessed by flow cytometry.Results: Pim-2 expression was enhanced in HCC. In post-transfected SMMC7721 cells, Pim-2 mRNA expression was up-regulated, level of Bad mRNA was attenuated, furthermore, the transcription level of Akt-1, SOCS-1, 4E-BP1, Bim and Puma gene wasn’t variety. Up-graulated Pim-2 can’t cause distinct change of cell cycle or apoptosis in hepatoma cell.Conclusions: The serine/threonine kinase Pim-2 plays an import role in the development of HCC, Pim-2 dependent maintenance of cell size and survival correlated with its ability to maintain down-regulated expression of the BH3 protein Bad. Pim-2 is not a trigger in cell-autonomous survival or inhibiting apoptosis in hepatocellular carcinoma. Pim-2 is a redundancy pathway of survival signaling.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 704-704
Author(s):  
Anjali Mishra ◽  
Shujun Liu ◽  
Gregory H Sams ◽  
Douglas P Curphey ◽  
Ramasamy Santhanam ◽  
...  

Abstract Abstract 704 Interleukin (IL)-15 is critical for the differentiation, proliferation, activation and survival of large granular lymphocytes (LGL). Malignant blasts from patients with acute LGL leukemia (LGLL) can express membrane bound IL-15 and often require IL-15 or IL-2 to survive and expand in vitro, suggesting a pivotal role of IL-15 in the genesis of LGLL in vivo. Indeed, 30% of mice engineered to over express IL-15 develop LGLL (J Exp Med 193:219-231, 2001), suggesting that IL-15 is a proto-oncogene. The present study examined the mechanism by which this may occur in mouse and in man. We observed ~2.5-fold increased levels of DNA methyltransferase 3b (Dnmt3b) in IL-15tg mice with LGLL compared to wild type (Wt) splenocytes (2.6 ± 0.6 -fold higher, N = 3 each, P =.03) and a ~2-fold increased levels of global DNA methylation (GDM) compared to Wt splenocytes (% global DNA levels measured by mass spec as % 5mC/(5mC+2dC): 3.6 ± 0.11%, N = 4 for IL-15tg LGLL; 1.5 ± 0.08%, N = 4 for Wt, P <.0001). Remarkably, samples from three LGLL patients exhibited significantly increased levels of DNMT3B when compared to their normal donor counterparts, ranging from 38-fold to 5448-fold (P <0.008), and a significant increase in GDM (4.4% ± 0.01 N = 3 for LGLL patients; 4.2 ± 0.01% N = 3 for normal donors, P =.008). Dnmt3b is negatively regulated by miR-29b, which is under the regulation of Myc (Nature Genetics 40:43-50, 2008). Blasts from both IL-15tg LGLL mice and LGLL patients had a significant decrease in miR-29b expression when compared to their Wt LGL and normal donor counterparts respectively (2-fold lower in mouse, P <.02; 11 to 230-fold lower in human samples, N = 3 each, P <.0009). This was accompanied by increased expression of Myc in LGLL samples (mouse LGLL: Wt LGL = 10.4 ± 1.3-fold higher, N = 3 each, P < 0.005; human LGLL: normal donor LGL = 7.7 ± 1.6 -fold higher, N = 4, P <.0015). Since IL-15 signaling appears important in development of LGLL we investigated the oncogenic effect of IL-15 by culturing sorted mouse Wt (NK1.1+) LGLs in medium supplemented with 10% FCS and 100 ng/ml IL-15. Within 12 hours of culture, these IL-15–stimulated Wt LGLs displayed 2.0 ± 0.5-fold lower levels of miR-29b due to increased binding of a repressor complex that included Myc in addition to NFkBp65 and Hdac1 at the miR-29b promoter (Fig. 1A). By day 30 of Wt LGL culture in IL-15, we observed a 2.6 ± 0.6-fold mRNA increase in Dnmt3b (N=3, P=.03) and a 10.4 ± 1.3-fold increase in Myc (N = 3, P < .005) compared to fresh Wt LGL, also confirmed at the protein level (Fig. 1B, green color). Compared to fresh Wt LGLs, day 30 IL-15-stimulated Wt LGLs exhibited a significant increase in GDM measured by mass spectrometry (%GDM: 2.5 ± 0.3 in stimulated Wt LGLs, N = 4, vs 1.5 ± 0.08 in non-stimulated Wt LGLs, N = 4, P <.01). Over several months, the IL-15-stimulated Wt LGLs continued to expand and accumulate centrosome aberrations and aneuploidy, with over expression of Aurora kinase B when compared to fresh Wt LGLs (170.5 ± 58.23 -fold higher, N = 3, P = .02). SCID mice injected with these transformed cells died of LGLL in the absence of exogenous IL-15. Thus, chronic in vitro exposure of Wt LGLs to excessive IL-15 results in leukemic transformation via a process that likely includes decreased miR-29b, increased Dnmt3b, global DNA hypermethylation, centrosome aberrations and chromosomal instability. To address the in vivo role of DNMT3B in IL-15-mediated leukemogenesis, we created DNMT3Btg mice under a vav-1 promoter. DNMT3Btg mice had normal survival at 2 years, while IL-15/DNMT3Btg mice showed a significantly lower latency and higher incidence of LGLL compared with IL-15tg mice (median survival: 20.7 wk in IL-15/DNMT3Btg vs 31.1 wk in IL-15tg, P <.0001; Fig. 2). Thirty-one of 42 (73%) IL-15/DNMT3Btg mice died of LGLL compared to 8 of 38 (21%) IL-15tg mice. Leukemic blasts from LGLL mice demonstrated excessive expression of Aurora kinase B over Wt splenocytes (257.7 ± 39.4 -fold higher, N = 3 each, P = .01) and aneuploidy. Collectively, we provide evidence that IL-15 is a proto-oncogene: excessive expression of this lymphocytotropic cytokine results in activation of a repressor complex that decreases expression of miR-29b, induces Dnmt3b expression, global DNA hypermethylation, aberrant centrosome formation, aneuploidy and leukemic transformation. We provide evidence for these mechanisms in both in vitro and in vivo mouse models as well as in primary LGLL patient samples. Disclosures: No relevant conflicts of interest to declare.


2017 ◽  
Author(s):  
Thanigaivelan Kanagasabai ◽  
Khalid Alhazzani ◽  
Thiagarajan Venkatesan ◽  
Sivanesan Dhandayuthapani ◽  
Ali Alaseem ◽  
...  

2018 ◽  
Author(s):  
Hiral Shah ◽  
Kanika Rawat ◽  
Harsh Ashar ◽  
Rajesh Patkar ◽  
Johannes Manjrekar

AbstractThe outer kinetochore DASH/DAM complex ensures proper spindle structure and chromosome segregation. While DASH complex protein requirement diverges among different yeasts, its role in filamentous fungi has not been investigated so far. We studied the dynamics and role of middle (Mis12) and outer (Dam1 and Ask1) kinetochore proteins in the filamentous fungal pathogen, Magnaporthe oryzae, which undergoes multiple cell cycle linked developmental transitions. Both Dam1 and Ask1, unlike Mis12, were recruited to the nucleus specifically during mitosis. While Dam1 was not required for viability, loss of its function (dam1Δ mutant) delayed mitotic progression, resulting in impaired conidial and hyphal development in Magnaporthe. Intriguingly, both Dam1 and Ask1 also localised to the hyphal tips, in the form of punctae oscillating back and forth from the growing ends, suggesting that Magnaporthe DASH complex proteins may play a non-canonical role in polarised growth during interphase, in addition to their function in nuclear segregation during mitosis. Impaired appressorial (infection structure) development and function in the dam1Δ mutant suggest that fungus-specific Dam1 complex proteins could be an attractive target for a novel anti-fungal strategy.Summary StatementDASH complex proteins are differentially recruited to the nucleus during cell division and are intriguingly involved in polarised growth during development and differentiation in the rice blast fungus.


Sign in / Sign up

Export Citation Format

Share Document