477 results on '"C-MYB"'
Search Results
2. Proto-oncogene c-Myb potentiates cisplatin resistance of ovarian cancer cells by downregulating lncRNA NKILA and modulating cancer stemness and LIN28A-let7 axis
- Author
-
Xue-Yan Zhang, Bo-Chi Zhu, Miao He, and Shan-Shan Dong
- Subjects
c-Myb ,NKILA ,LIN28A ,Let-7 ,Cisplatin resistance ,Ovarian cancer ,Gynecology and obstetrics ,RG1-991 - Abstract
Abstract Ovarian cancer is a major gynecological cancer that has poor prognosis associated mainly to its late diagnosis. Cisplatin is an FDA approved ovarian cancer therapy and even though the therapy is initially promising, the patients mostly progress to resistance against cisplatin. The underlying mechanisms are complex and not very clearly understood. Using two different paired cell lines representing cisplatin-sensitive and the cisplatin-resistant ovarian cancer cells, the ES2 and the A2780 parental and cisplatin-resistant cells, we show an elevated proto-oncogene c-Myb in resistant cells. We further show down-regulated lncRNA NKILA in resistant cells with its de-repression in resistant cells when c-Myb is silenced. NKILA negatively correlates with cancer cell and invasion but has no effect on cellular proliferation or cell cycle. C-Myb activates NF-κB signaling which is inhibited by NKILA. The cisplatin resistant cells are also marked by upregulated stem cell markers, particularly LIN28A and OCT4, and downregulated LIN28A-targeted let-7 family miRNAs. Whereas LIN28A and downregulated let-7s individually de-repress c-Myb-mediated cisplatin resistance, the ectopic expression of let-7s attenuates LIN28A effects, thus underlying a c-Myb-NKILA-LIN28A-let-7 axis in cisplatin resistance of ovarian cancer cells that needs to be further explored for therapeutic intervention.
- Published
- 2024
- Full Text
- View/download PDF
3. c‐myb is involved in CML progression and is a therapeutic target in the zebrafish CML model
- Author
-
Yin Ye, Xiaojun Yang, Feifei Li, Wei Liu, Wenqing Zhang, and Zhibin Huang
- Subjects
chronic myeloid leukemia ,c‐myb ,flavopiridol ,zebrafish model ,Medicine (General) ,R5-920 - Abstract
Abstract Background Despite the success of tyrosine kinase inhibitors in chronic myeloid leukemia (CML) therapy, CML still faces the challenges of drug resistance and progression to blast crisis. Twenty‐five percent of patients have imatinib resistance and treatment difficulties due to heterogeneity after progression, but little is known about the mechanism. A key transcription factor in hematopoiesis, MYB, has been reported to increase abnormally in several types of aggressive blood disorders including CML. Methods This study used a zebrafish model to explore the relationship between BCR/ABL1 and c‐myb in CML progression. A CML zebrafish model was crossed with a c‐myb hyperactivity transgenic line. Results It was found that both exogenous BCR/ABL1 and c‐myb could up‐regulate the expression of neutrophil‐related genes. More seriously, neutrophil accumulation was observed when BCR/ABL1 was combined with c‐myb overexpression. Further studies showed that c‐myb may be one of the downstream targets of BCR/ABL1 and the effect of BCR/ABL1 on neutrophils was c‐myb dependent. Taking advantage of this inheritable in vivo model, it was shown that a combination of imatinib and flavopiridol, a cyclin‐dependent kinase inhibitor targeting MYB, could more effectively alleviate the aggressive phenotype of the double transgene line. Conclusion In summary, this study suggests that c‐myb acts downstream of BCR/ABL1 and is involved in CML progression and is therefore a risk factor and a valuable target for the treatment of CML progression. The model used in the study could be helpful in high‐throughput drug screening in CML transformation.
- Published
- 2024
- Full Text
- View/download PDF
4. Proto-oncogene c-Myb potentiates cisplatin resistance of ovarian cancer cells by downregulating lncRNA NKILA and modulating cancer stemness and LIN28A-let7 axis
- Author
-
Zhang, Xue-Yan, Zhu, Bo-Chi, He, Miao, and Dong, Shan-Shan
- Published
- 2024
- Full Text
- View/download PDF
5. Multiphoton In Vivo Microscopy of Embryonic Thrombopoiesis Reveals the Generation of Platelets through Budding.
- Author
-
Liu, Huan, Ishikawa-Ankerhold, Hellen, Winterhalter, Julia, Lorenz, Michael, Vladymyrov, Mykhailo, Massberg, Steffen, Schulz, Christian, and Orban, Mathias
- Subjects
- *
BLOOD platelets , *YOLK sac , *MICROSCOPY , *LABORATORY mice , *MEGAKARYOCYTES , *THROMBOPOIETIN receptors , *COINCIDENCE - Abstract
Platelets are generated by specialized cells called megakaryocytes (MKs). However, MK's origin and platelet release mode have remained incompletely understood. Here, we established direct visualization of embryonic thrombopoiesis in vivo by combining multiphoton intravital microscopy (MP-IVM) with a fluorescence switch reporter mouse model under control of the platelet factor 4 promoter (Pf4CreRosa26mTmG). Using this microscopy tool, we discovered that fetal liver MKs provide higher thrombopoietic activity than yolk sac MKs. Mechanistically, fetal platelets were released from MKs either by membrane buds or the formation of proplatelets, with the former constituting the key process. In E14.5 c-Myb-deficient embryos that lack definitive hematopoiesis, MK and platelet numbers were similar to wild-type embryos, indicating the independence of embryonic thrombopoiesis from definitive hematopoiesis at this stage of development. In summary, our novel MP-IVM protocol allows the characterization of thrombopoiesis with high spatio-temporal resolution in the mouse embryo and has identified membrane budding as the main mechanism of fetal platelet production. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. Molecular dynamics simulation reveals DNA-specific recognition mechanism via c-Myb in pseudo-palindromic consensus of mim-1 promoter.
- Author
-
Weng, Jinru, Yang, Shuo, Shen, Jinkang, Liu, Hongsen, Xu, Yuzi, Hao, Dongyun, and Wang, Shan
- Abstract
Copyright of Journal of Zhejiang University: Science B is the property of Springer Nature and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2023
- Full Text
- View/download PDF
7. rs71327024 Associated with COVID-19 Hospitalization Reduces CXCR6 Promoter Activity in Human CD4 + T Cells via Disruption of c-Myb Binding.
- Author
-
Uvarova, Aksinya N., Stasevich, Ekaterina M., Ustiugova, Alina S., Mitkin, Nikita A., Zheremyan, Elina A., Sheetikov, Savely A., Zornikova, Ksenia V., Bogolyubova, Apollinariya V., Rubtsov, Mikhail A., Kulakovskiy, Ivan V., Kuprash, Dmitry V., Korneev, Kirill V., and Schwartz, Anton M.
- Subjects
- *
T cells , *T helper cells , *COVID-19 , *BINDING sites , *CD4 antigen , *T cell receptors , *CHEMOKINE receptors - Abstract
Single-nucleotide polymorphism rs71327024 located in the human 3p21.31 locus has been associated with an elevated risk of hospitalization upon SARS-CoV-2 infection. The 3p21.31 locus contains several genes encoding chemokine receptors potentially relevant to severe COVID-19. In particular, CXCR6, which is prominently expressed in T lymphocytes, NK, and NKT cells, has been shown to be involved in the recruitment of immune cells to non-lymphoid organs in chronic inflammatory and respiratory diseases. In COVID-19, CXCR6 expression is reduced in lung resident memory T cells from patients with severe disease as compared to the control cohort with moderate symptoms. We demonstrate here that rs71327024 is located within an active enhancer that augments the activity of the CXCR6 promoter in human CD4+ T lymphocytes. The common rs71327024(G) variant makes a functional binding site for the c-Myb transcription factor, while the risk rs71327024(T) variant disrupts c-Myb binding and reduces the enhancer activity. Concordantly, c-Myb knockdown in PMA-treated Jurkat cells negates rs71327024's allele-specific effect on CXCR6 promoter activity. We conclude that a disrupted c-Myb binding site may decrease CXCR6 expression in T helper cells of individuals carrying the minor rs71327024(T) allele and thus may promote the progression of severe COVID-19 and other inflammatory pathologies. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
8. Circular RNA circLOC101928570 suppresses systemic lupus erythematosus progression by targeting the miR-150-5p/c-myb axis
- Author
-
Xingwang Zhao, Rui Dong, Zhongwei Tang, Juan Wang, Chunyou Wang, Zhiqiang Song, Bing Ni, Longlong Zhang, Xiaochong He, and Yi You
- Subjects
Systemic lupus erythematosus ,circLOC101928570 ,miR-150-5p ,c-myb ,Biomarker ,Medicine - Abstract
Abstract Background Accumulating evidence supports the implication of circular RNAs (circRNAs) in systemic lupus erythematosus (SLE). However, little is known about the detailed mechanisms and roles of circRNAs in the pathogenesis of SLE. Methods Quantitative real-time PCR was used to determine the levels of circLOC101928570 and miR-150-5p in peripheral blood mononuclear cells of SLE. Overexpression and knockdown experiments were conducted to assess the effects of circLOC101928570. Fluorescence in situ hybridization, RNA immunoprecipitation, luciferase reporter assays, Western blot, flow cytometry analysis and enzyme-linked immunosorbent assay were used to investigate the molecular mechanisms underlying the function of circLOC101928570. Results The results showed that the level of circLOC101928570 was significantly downregulated in SLE and correlated with the systemic lupus erythematosus disease activity index. Functionally, circLOC101928570 acted as a miR-150-5p sponge to relieve the repressive effect on its target c-myb, which modulates the activation of immune inflammatory responses. CircLOC101928570 knockdown enhanced apoptosis. Moreover, circLOC101928570 promoted the transcriptional level of IL2RA by directly regulating the miR-150-5p/c-myb axis. Conclusion Overall, our findings demonstrated that circLOC101928570 played a critical role in SLE. The downregulation of circLOC101928570 suppressed SLE progression through the miR-150-5p/c-myb/IL2RA axis. Our findings identified that circLOC101928570 serves as a potential biomarker for the diagnosis and therapy of SLE.
- Published
- 2022
- Full Text
- View/download PDF
9. Multiphoton In Vivo Microscopy of Embryonic Thrombopoiesis Reveals the Generation of Platelets through Budding
- Author
-
Huan Liu, Hellen Ishikawa-Ankerhold, Julia Winterhalter, Michael Lorenz, Mykhailo Vladymyrov, Steffen Massberg, Christian Schulz, and Mathias Orban
- Subjects
megakaryocyte ,multiphoton intravital microscopy ,thrombopoiesis ,yolk sac ,fetal liver ,c-Myb ,Cytology ,QH573-671 - Abstract
Platelets are generated by specialized cells called megakaryocytes (MKs). However, MK’s origin and platelet release mode have remained incompletely understood. Here, we established direct visualization of embryonic thrombopoiesis in vivo by combining multiphoton intravital microscopy (MP-IVM) with a fluorescence switch reporter mouse model under control of the platelet factor 4 promoter (Pf4CreRosa26mTmG). Using this microscopy tool, we discovered that fetal liver MKs provide higher thrombopoietic activity than yolk sac MKs. Mechanistically, fetal platelets were released from MKs either by membrane buds or the formation of proplatelets, with the former constituting the key process. In E14.5 c-Myb-deficient embryos that lack definitive hematopoiesis, MK and platelet numbers were similar to wild-type embryos, indicating the independence of embryonic thrombopoiesis from definitive hematopoiesis at this stage of development. In summary, our novel MP-IVM protocol allows the characterization of thrombopoiesis with high spatio-temporal resolution in the mouse embryo and has identified membrane budding as the main mechanism of fetal platelet production.
- Published
- 2023
- Full Text
- View/download PDF
10. rs71327024 Associated with COVID-19 Hospitalization Reduces CXCR6 Promoter Activity in Human CD4+ T Cells via Disruption of c-Myb Binding
- Author
-
Aksinya N. Uvarova, Ekaterina M. Stasevich, Alina S. Ustiugova, Nikita A. Mitkin, Elina A. Zheremyan, Savely A. Sheetikov, Ksenia V. Zornikova, Apollinariya V. Bogolyubova, Mikhail A. Rubtsov, Ivan V. Kulakovskiy, Dmitry V. Kuprash, Kirill V. Korneev, and Anton M. Schwartz
- Subjects
3p21.31 locus ,COVID-19 ,CXCR6 ,T helpers ,c-Myb ,non-coding SNP ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Single-nucleotide polymorphism rs71327024 located in the human 3p21.31 locus has been associated with an elevated risk of hospitalization upon SARS-CoV-2 infection. The 3p21.31 locus contains several genes encoding chemokine receptors potentially relevant to severe COVID-19. In particular, CXCR6, which is prominently expressed in T lymphocytes, NK, and NKT cells, has been shown to be involved in the recruitment of immune cells to non-lymphoid organs in chronic inflammatory and respiratory diseases. In COVID-19, CXCR6 expression is reduced in lung resident memory T cells from patients with severe disease as compared to the control cohort with moderate symptoms. We demonstrate here that rs71327024 is located within an active enhancer that augments the activity of the CXCR6 promoter in human CD4+ T lymphocytes. The common rs71327024(G) variant makes a functional binding site for the c-Myb transcription factor, while the risk rs71327024(T) variant disrupts c-Myb binding and reduces the enhancer activity. Concordantly, c-Myb knockdown in PMA-treated Jurkat cells negates rs71327024’s allele-specific effect on CXCR6 promoter activity. We conclude that a disrupted c-Myb binding site may decrease CXCR6 expression in T helper cells of individuals carrying the minor rs71327024(T) allele and thus may promote the progression of severe COVID-19 and other inflammatory pathologies.
- Published
- 2023
- Full Text
- View/download PDF
11. Circular RNA circLOC101928570 suppresses systemic lupus erythematosus progression by targeting the miR-150-5p/c-myb axis.
- Author
-
Zhao, Xingwang, Dong, Rui, Tang, Zhongwei, Wang, Juan, Wang, Chunyou, Song, Zhiqiang, Ni, Bing, Zhang, Longlong, He, Xiaochong, and You, Yi
- Subjects
- *
CIRCULAR RNA , *SYSTEMIC lupus erythematosus , *MONONUCLEAR leukocytes , *FLUORESCENCE in situ hybridization , *ENZYME-linked immunosorbent assay - Abstract
Background: Accumulating evidence supports the implication of circular RNAs (circRNAs) in systemic lupus erythematosus (SLE). However, little is known about the detailed mechanisms and roles of circRNAs in the pathogenesis of SLE.Methods: Quantitative real-time PCR was used to determine the levels of circLOC101928570 and miR-150-5p in peripheral blood mononuclear cells of SLE. Overexpression and knockdown experiments were conducted to assess the effects of circLOC101928570. Fluorescence in situ hybridization, RNA immunoprecipitation, luciferase reporter assays, Western blot, flow cytometry analysis and enzyme-linked immunosorbent assay were used to investigate the molecular mechanisms underlying the function of circLOC101928570.Results: The results showed that the level of circLOC101928570 was significantly downregulated in SLE and correlated with the systemic lupus erythematosus disease activity index. Functionally, circLOC101928570 acted as a miR-150-5p sponge to relieve the repressive effect on its target c-myb, which modulates the activation of immune inflammatory responses. CircLOC101928570 knockdown enhanced apoptosis. Moreover, circLOC101928570 promoted the transcriptional level of IL2RA by directly regulating the miR-150-5p/c-myb axis.Conclusion: Overall, our findings demonstrated that circLOC101928570 played a critical role in SLE. The downregulation of circLOC101928570 suppressed SLE progression through the miR-150-5p/c-myb/IL2RA axis. Our findings identified that circLOC101928570 serves as a potential biomarker for the diagnosis and therapy of SLE. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
12. Conformation dynamics of the intrinsically disordered protein c-Myb with the ff99IDPs force field
- Author
-
Guo, Xiang, Han, Jincheng, Luo, Ray, and Chen, Hai-Feng
- Subjects
Chemical Sciences ,Theoretical and Computational Chemistry ,Emerging Infectious Diseases ,1.1 Normal biological development and functioning ,Underpinning research ,Generic health relevance ,c-Myb ,conformation dynamics ,dynamics correlation network ,ff99IDPs force field ,solvent model ,Chemical sciences - Abstract
The intrinsically disordered protein c-Myb plays a critical role in cellular proliferation and differentiation. Loss of c-myb function results in embryonic lethality due to failure of fetal hepatic hematopoiesis. The conformation dynamics of the intrinsically disordered c-Myb are still unknown. Here, molecular dynamics (MD) simulations with the intrinsically disordered protein force field ff99IDPs were used to study the conformation dynamics. In comparison with ff99SBildn, ff99IDPs can reproduce more diverse disordered conformers of c-Myb. The predicted secondary chemical shift under ff99IDPs is more close to that of experiment data than that under ff99SBildn. Therefore, ff99IDPs can sample native molten globule, native pre-molten globule and native coil conformers for c-Myb. These results are consistent with those of other intrinsically disordered proteins. Kinetic analysis of MD simulations shows that c-Myb folds via a two-state process and indicates that c-Myb folds in the order of tertiary folding and helical folding. The folding nucleus of KEL plays an essential role in stabilizing the folding state with dynamic correlation networks. The influences of solvent models for TIP3P, TIP4P-EW and TIP5P were also investigated and it was found that TIP3P and ff99IDPs are the best combination to research the conformer sampling of c-Myb. These results reveal the conformation dynamics of c-Myb and confirm that the ff99IDPs force field can be used to research the relationship between structure and function of other intrinsically disordered proteins.
- Published
- 2017
13. c-Myb interferes with inflammatory IL1α-NF-κB pathway in breast cancer cells
- Author
-
Monika Dúcka, Martina Kučeríková, Filip Trčka, Jakub Červinka, Elisabetta Biglieri, Jan Šmarda, Lubor Borsig, Petr Beneš, and Lucia Knopfová
- Subjects
Breast cancer ,c-Myb ,IL1α ,NF-κB ,Inflammation ,Transactivation ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The transcription factor c-Myb can be involved in the activation of many genes with protumorigenic function; however, its role in breast cancer (BC) development is still under discussion. c-Myb is considered as a tumor-promoting factor in the early phases of BC, on the other hand, its expression in BC patients relates to a good prognosis. Previously, we have shown that c-Myb controls the capacity of BC cells to form spontaneous lung metastasis. Reduced seeding of BC cells to the lungs is linked to high expression of c-Myb and a decline in expression of a specific set of inflammatory genes. Here, we unraveled a c-Myb-IL1α-NF-κB signaling axis that takes place in tumor cells. We report that an overexpression of c-Myb interfered with the activity of NF-κB in several BC cell lines. We identified IL1α to be essential for this interference since it was abrogated in the IL1α-deficient cells. Overexpression of IL1α, as well as addition of recombinant IL1α protein, activated NF-κB signaling and restored expression of the inflammatory signature genes suppressed by c-Myb. The endogenous levels of c-Myb negatively correlated with IL1α on both transcriptional and protein levels across BC cell lines. We concluded that inhibition of IL1α expression by c-Myb reduces NF-κB activity and disconnects the inflammatory circuit, a potentially targetable mechanism to mimic the antimetastatic effect of c-Myb with therapeutic perspective.
- Published
- 2021
- Full Text
- View/download PDF
14. Transcription factor c-Myb: novel prognostic factor in osteosarcoma.
- Author
-
Říhová, Kamila, Dúcka, Monika, Zambo, Iva Staniczková, Vymětalová, Ladislava, Šrámek, Martin, Trčka, Filip, Verner, Jan, Drápela, Stanislav, Fedr, Radek, Suchánková, Tereza, Pavlatovská, Barbora, Ondroušková, Eva, Kubelková, Irena, Zapletalová, Danica, Tuček, Štěpán, Múdry, Peter, Krákorová, Dagmar Adámková, Knopfová, Lucia, Šmarda, Jan, and Souček, Karel
- Abstract
The transcription factor c-Myb is an oncoprotein promoting cell proliferation and survival when aberrantly activated/expressed, thus contributing to malignant transformation. Overexpression of c-Myb has been found in leukemias, breast, colon and adenoid cystic carcinoma. Recent studies revealed its expression also in osteosarcoma cell lines and suggested its functional importance during bone development. However, the relevance of c-Myb in control of osteosarcoma progression remains unknown. A retrospective clinical study was carried out to assess a relationship between c-Myb expression in archival osteosarcoma tissues and prognosis in a cohort of high-grade osteosarcoma patients. In addition, MYB was depleted in metastatic osteosarcoma cell lines SAOS-2 LM5 and 143B and their growth, chemosensitivity, migration and metastatic activity were determined. Immunohistochemical analysis revealed that high c-Myb expression was significantly associated with poor overall survival in the cohort and metastatic progression in young patients. Increased level of c-Myb was detected in metastatic osteosarcoma cell lines and its depletion suppressed their growth, colony-forming capacity, migration and chemoresistance in vitro in a cell line-dependent manner. MYB knock-out resulted in reduced metastatic activity of both SAOS-2 LM5 and 143B cell lines in immunodeficient mice. Transcriptomic analysis revealed the c-Myb-driven functional programs enriched for genes involved in the regulation of cell growth, stress response, cell adhesion and cell differentiation/morphogenesis. Wnt signaling pathway was identified as c-Myb target in osteosarcoma cells. Taken together, we identified c-Myb as a negative prognostic factor in osteosarcoma and showed its involvement in the regulation of osteosarcoma cell growth, chemosensitivity, migration and metastatic activity. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
15. Destabilization of macrophage migration inhibitory factor by 4‐IPP reduces NF‐κB/P‐TEFb complex‐mediated c‐Myb transcription to suppress osteosarcoma tumourigenesis.
- Author
-
Zheng, Lin, Feng, Zhenhua, Tao, Siyue, Gao, Jiawei, Lin, Ye, Wei, Xiaoan, Zheng, Bingjie, Huang, Bao, Zheng, Zeyu, Zhang, Xuyang, Liu, Junhui, Shan, Zhi, Chen, Yilei, Chen, Jian, and Zhao, Fengdong
- Subjects
- *
PROTEOLYSIS , *MACROPHAGE migration inhibitory factor , *OSTEOSARCOMA , *ONCOGENIC proteins - Abstract
Background: As an inflammatory factor and oncogenic driver protein, the pleiotropic cytokine macrophage migration inhibitory factor (MIF) plays a crucial role in the osteosarcoma microenvironment. Although 4‐iodo‐6‐phenylpyrimidine (4‐IPP) can inactivate MIF biological functions, its anti‐osteosarcoma effect and molecular mechanisms have not been investigated. In this study, we identified the MIF inhibitor 4‐IPP as a specific double‐effector drug for osteosarcoma with both anti‐tumour and anti‐osteoclastogenic functions. Methods: The anti‐cancer effects of 4‐IPP were evaluated by wound healing assay, cell cycle analysis, colony formation assay, CCK‐8 assay, apoptosis analysis, and Transwell migration/invasion assays. Through the application of a luciferase reporter, chromatin immunoprecipitation assays, and immunofluorescence and coimmunoprecipitation analyses, the transcriptional regulation of the NF‐κB/P‐TEFb complex on c‐Myb‐ and STUB1‐mediated proteasome‐dependent MIF protein degradation was confirmed. The effect of 4‐IPP on tumour growth and metastasis was assessed using an HOS‐derived tail vein metastasis model and subcutaneous and orthotopic xenograft tumour models. Results: In vitro, 4‐IPP significantly reduced the proliferation and metastasis of osteosarcoma cells by suppressing the NF‐κB pathway. 4‐IPP hindered the binding between MIF and CD74 as well as p65. Moreover, 4‐IPP inhibited MIF to interrupt the formation of downstream NF‐κB/P‐TEFb complexes, leading to the down‐regulation of c‐Myb transcription. Interestingly, the implementation of 4‐IPP can mediate small molecule‐induced MIF protein proteasomal degradation via the STUB1 E3 ligand. However, 4‐IPP still interrupted MIF‐mediated communication between osteosarcoma cells and osteoclasts, thus promoting osteoclastogenesis. Remarkably, 4‐IPP strongly reduced HOS‐derived xenograft osteosarcoma tumourigenesis and metastasis in an in vivo mouse model. Conclusions: Our findings demonstrate that the small molecule 4‐IPP targeting the MIF protein exerts an anti‐osteosarcoma effect by simultaneously inactivating the biological functions of MIF and promoting its proteasomal degradation. Direct destabilization of the MIF protein with 4‐IPP may be a promising therapeutic strategy for treating osteosarcoma. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
16. Dissecting the transactivation domain (tAD) of the transcription factor c‐Myb to assess recent models of tAD function
- Author
-
Guro Næs, Jan Ove Storesund, Priyanga‐Dina Udayakumar, Marit Ledsaak, and Odd Stokke Gabrielsen
- Subjects
chromatin ,c‐Myb ,transactivation domain ,transcription factor ,yeast ,Biology (General) ,QH301-705.5 - Abstract
Transcription factors use a DNA‐binding domain to localize their action and a transactivation domain (tAD) to stimulate activation of the associated gene. Recent work has renewed interest in how tADs activate genes, which remains poorly understood. Key features in the new models are exposure of short linear motifs (SLMs) and liquid–liquid phase separation (LLPS). Inspired by the new models for tAD function, we decided to revisit the tAD of the haematopoietic transcription factor c‐Myb by performing a mutational analysis to see how these new models fit and potentially explain the tAD behaviour of this master regulator. We know that c‐Myb has an acidic tAD, which contains a well‐characterized SLM in the form of a LxxLL motif. By testing 12 alanine‐scanning mutants and three mutants with major reorganization of its tAD in two mammalian reporter systems, we found a pattern of effects very close to what would be expected from the SLM‐exposure model, with strong effects exerted by both acidic replacements and SLM mutation. When the same mutants were tested in a yeast system, the pattern of effects was dramatically different, with the SLM mutation exerting no effect, and tAD behaviour was much less affected by small alterations, as would be expected from a LLPS model. These observations are discussed in the light of the two new tAD models, and a two‐step hypothesis for transactivation, combining both models, is proposed.
- Published
- 2020
- Full Text
- View/download PDF
17. Destabilization of macrophage migration inhibitory factor by 4‐IPP reduces NF‐κB/P‐TEFb complex‐mediated c‐Myb transcription to suppress osteosarcoma tumourigenesis
- Author
-
Lin Zheng, Zhenhua Feng, Siyue Tao, Jiawei Gao, Ye Lin, Xiaoan Wei, Bingjie Zheng, Bao Huang, Zeyu Zheng, Xuyang Zhang, Junhui Liu, Zhi Shan, Yilei Chen, Jian Chen, and Fengdong Zhao
- Subjects
4‐IPP ,CDK9 ,c‐Myb ,MIF ,osteosarcoma ,STUB1 ,Medicine (General) ,R5-920 - Abstract
Abstract Background As an inflammatory factor and oncogenic driver protein, the pleiotropic cytokine macrophage migration inhibitory factor (MIF) plays a crucial role in the osteosarcoma microenvironment. Although 4‐iodo‐6‐phenylpyrimidine (4‐IPP) can inactivate MIF biological functions, its anti‐osteosarcoma effect and molecular mechanisms have not been investigated. In this study, we identified the MIF inhibitor 4‐IPP as a specific double‐effector drug for osteosarcoma with both anti‐tumour and anti‐osteoclastogenic functions. Methods The anti‐cancer effects of 4‐IPP were evaluated by wound healing assay, cell cycle analysis, colony formation assay, CCK‐8 assay, apoptosis analysis, and Transwell migration/invasion assays. Through the application of a luciferase reporter, chromatin immunoprecipitation assays, and immunofluorescence and coimmunoprecipitation analyses, the transcriptional regulation of the NF‐κB/P‐TEFb complex on c‐Myb‐ and STUB1‐mediated proteasome‐dependent MIF protein degradation was confirmed. The effect of 4‐IPP on tumour growth and metastasis was assessed using an HOS‐derived tail vein metastasis model and subcutaneous and orthotopic xenograft tumour models. Results In vitro, 4‐IPP significantly reduced the proliferation and metastasis of osteosarcoma cells by suppressing the NF‐κB pathway. 4‐IPP hindered the binding between MIF and CD74 as well as p65. Moreover, 4‐IPP inhibited MIF to interrupt the formation of downstream NF‐κB/P‐TEFb complexes, leading to the down‐regulation of c‐Myb transcription. Interestingly, the implementation of 4‐IPP can mediate small molecule‐induced MIF protein proteasomal degradation via the STUB1 E3 ligand. However, 4‐IPP still interrupted MIF‐mediated communication between osteosarcoma cells and osteoclasts, thus promoting osteoclastogenesis. Remarkably, 4‐IPP strongly reduced HOS‐derived xenograft osteosarcoma tumourigenesis and metastasis in an in vivo mouse model. Conclusions Our findings demonstrate that the small molecule 4‐IPP targeting the MIF protein exerts an anti‐osteosarcoma effect by simultaneously inactivating the biological functions of MIF and promoting its proteasomal degradation. Direct destabilization of the MIF protein with 4‐IPP may be a promising therapeutic strategy for treating osteosarcoma.
- Published
- 2022
- Full Text
- View/download PDF
18. c‐Myb facilitates immune escape of esophageal adenocarcinoma cells through the miR‐145‐5p/SPOP/PD‐L1 axis.
- Author
-
Zhang, Lan, Wang, Xiaohui, Li, Yunfei, Han, Jing, Gao, Xianzheng, Li, Shenglei, and Wang, Feng
- Subjects
- *
LABORATORY mice , *ADENOCARCINOMA , *DEATH rate , *CELL migration , *PROGNOSIS , *T cells - Abstract
Esophageal adenocarcinoma (EAC), a subtype of esophageal carcinoma, is a severe health problem associated with high death rate and poor prognosis. Immunotherapy has proven to be effective in many solid tumors, including EAC, but immune escape blocks its effectiveness. Thus, we explored the mechanisms and functional role of c‐Myb in immune escape of EAC cells. Clinical EAC tissues were collected for determining the expression of c‐Myb, speckled POZ protein (SPOP), and miR‐145‐5p. Functional assays were then performed to detect the interactions between c‐Myb and SPOP as well as between SPOP and miR‐145‐5p. EAC cell invasion and migration were assessed. Next, T cells were sorted and cocultured with EAC cells with different treatments followed by detection of T‐cell viability. In addition, a mouse model of EAC was constructed for relevant in vivo assays. c‐Myb and miR‐145‐5p were highly expressed and SPOP had low expressions in EAC. c‐Myb activated the transcription of miR‐145‐5p, which in turn targeted SPOP. Further, SPOP accelerated the ubiquitination of PD‐L1 to enhance its expression. Overexpression of PD‐L1 suppressed T‐cell functions and promoted proliferative and migrative abilities of EAC cells to induce immune escape. The above findings were also confirmed in the ECA mouse model in vivo. Our findings uncovered that c‐Myb can promote the immune escape of EAC cells by favoring the transcription of miR‐145‐5p and inhibiting SPOP‐dependent ubiquitination and degradation of PD‐L1, thus, presenting new target for EAC adjunct therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
19. c‐Myb facilitates immune escape of esophageal adenocarcinoma cells through the miR‐145‐5p/SPOP/PD‐L1 axis
- Author
-
Lan Zhang, Xiaohui Wang, Yunfei Li, Jing Han, Xianzheng Gao, Shenglei Li, and Feng Wang
- Subjects
c‐Myb ,esophageal adenocarcinoma ,immune escape ,microRNA‐145‐5p ,programmed death ligand 1 ,speckled POZ protein ,Medicine (General) ,R5-920 - Abstract
Abstract Esophageal adenocarcinoma (EAC), a subtype of esophageal carcinoma, is a severe health problem associated with high death rate and poor prognosis. Immunotherapy has proven to be effective in many solid tumors, including EAC, but immune escape blocks its effectiveness. Thus, we explored the mechanisms and functional role of c‐Myb in immune escape of EAC cells. Clinical EAC tissues were collected for determining the expression of c‐Myb, speckled POZ protein (SPOP), and miR‐145‐5p. Functional assays were then performed to detect the interactions between c‐Myb and SPOP as well as between SPOP and miR‐145‐5p. EAC cell invasion and migration were assessed. Next, T cells were sorted and cocultured with EAC cells with different treatments followed by detection of T‐cell viability. In addition, a mouse model of EAC was constructed for relevant in vivo assays. c‐Myb and miR‐145‐5p were highly expressed and SPOP had low expressions in EAC. c‐Myb activated the transcription of miR‐145‐5p, which in turn targeted SPOP. Further, SPOP accelerated the ubiquitination of PD‐L1 to enhance its expression. Overexpression of PD‐L1 suppressed T‐cell functions and promoted proliferative and migrative abilities of EAC cells to induce immune escape. The above findings were also confirmed in the ECA mouse model in vivo. Our findings uncovered that c‐Myb can promote the immune escape of EAC cells by favoring the transcription of miR‐145‐5p and inhibiting SPOP‐dependent ubiquitination and degradation of PD‐L1, thus, presenting new target for EAC adjunct therapy.
- Published
- 2021
- Full Text
- View/download PDF
20. c-Myb
- Author
-
Ness, Scott A. and Choi, Sangdun, editor
- Published
- 2018
- Full Text
- View/download PDF
21. CURE 2000
- Author
-
Iversen, Patrick L. and Iversen, Patrick L.
- Published
- 2018
- Full Text
- View/download PDF
22. lncRNA SNHG4 inhibits ferroptosis by orchestrating miR-150-5p/c-Myb axis in colorectal cancer.
- Author
-
Li, Si-qi, Lv, Feng, Xu, Wen-ting, Yin, Yi-xin, Wei, Hao-tang, Li, Ke-zhi, and Hu, Bang-li
- Subjects
- *
COLORECTAL cancer , *GENE expression , *LINCRNA , *GENETIC overexpression , *ANIMAL models in research - Abstract
LncRNAs have shown to regulate ferroptosis in colorectal cancer (CRC), but the mechanism remains largely unknown. This study unveiled the mechanism of SNHG4 underlying ferroptosis in CRC. RNA-seq and RT-PCR assay confirmed SNHG4 was decreased after Erastin treatment in CRC cells. Overexpression of SNHG4 inhibited and silence promoted CRC cells ferroptosis. SNHG4 was positively correlated to c-Myb in CRC tissues and both located in cytoplasm of CRC cells. RIP and RNA pull-down assays verified the interaction between SNHG4 and c-Myb. Silence of c-Myb alleviated the suppressing effect on ferroptosis by SNHG4 in CRC cells. Dual-luciferase reporter assay revealed that SNHG4 sponging miR-150-5p in CRC cells. Overexpression of SNHG4 decreased the miR-150-5p and increased c-Myb expression. c-Myb was a direct target gene of miR-150-5p in CRC cells. Moreover, effect of CDO1 on ferroptosis was regulated transcriptionally by c-Myb, overexpression of c-Myb reduce CDO1 expression and enhance the GPX4 levels. The animal models confirmed that regulatory effect of SNHG4 on miR-150-5p and c-Myb after inducing ferroptosis. We concluded that SNHG4 inhibited Erastin-induce ferroptosis in CRC, this effect is via sponging miR-150-5p to regulate c-Myb expression, and activated CDO1/GPX4 axis. These findings provide insights into the regulatory mechanism of SNHG4 on ferroptosis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
23. c‐MYB‐ and PGC1a‐dependent metabolic switch induced by MYBBP1A loss in renal cancer
- Author
-
Blanca Felipe‐Abrio, Eva M. Verdugo‐Sivianes, and Amancio Carnero
- Subjects
c‐MYB ,metabolism ,MYBBP1A ,PGC1α ,renal cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The tumor microenvironment may alter the original tumorigenic potential of tumor cells. Under harsh environmental conditions, genetic alterations conferring selective advantages may initiate the growth of tumor subclones, providing new opportunities for these tumors to grow. We performed a genetic loss‐of‐function screen to identify genetic alterations able to promote tumor cell growth in the absence of glucose. We identified that downregulation of MYBBP1A increases tumorigenic properties under nonpermissive conditions. MYBBP1A downregulation simultaneously activates PGC1α, directly by alleviating direct repression and indirectly by increasing PGC1α mRNA levels through c‐MYB, leading to a metabolic switch from glycolysis to OXPHOS and increased tumorigenesis in low‐glucose microenvironments. We have also identified reduced MYBBP1A expression in human renal tumor samples, which show high expression levels of genes involved in oxidative metabolism. In summary, our data support the role of MYBBP1A as a tumor suppressor by regulating c‐MYB and PGC1α. Therefore, loss of MYBBP1A increases adaptability spanning of tumors through metabolic switch.
- Published
- 2019
- Full Text
- View/download PDF
24. Analysis of the FBXO7 promoter reveals overlapping Pax5 and c-Myb binding sites functioning in B cells.
- Author
-
Harris, Rebecca, Randle, Suzanne, and Laman, Heike
- Subjects
- *
BINDING sites , *B cells , *CELL physiology , *TRANSCRIPTION factors , *PROMOTERS (Genetics) - Abstract
Fbxo7 is a key player in the differentiation and function of numerous blood cell types, and in neurons, oligodendrocytes and spermatocytes. In an effort to gain insight into the physiological and pathological settings where Fbxo7 is likely to play a key role, we sought to define the transcription factors which direct FBXO7 expression. Using sequence alignments across 28 species, we defined the human FBXO7 promoter and found that it contains two conserved regions enriched for multiple transcription factor binding sites. Many of these have roles in either neuronal or haematopoietic development. Using various FBXO7 promoter reporters, we found ELF4, Pax5 and c-Myb have functional binding sites that activate transcription. We find endogenous Pax5 is bound to the FBXO7 promoter in pre-B cells, and that the exogenous expression of Pax5 represses Fbxo7 transcription in early pro-B cells. • We defined the human FBXO7 promoter, as a conserved promoter region between −1300 and + 100 bp from the start of exon 1. • Two conserved islands of putative transcription factor binding sites were found with 32 putative binding sites identified for 24 different TFs (17 in the distal region; 15 in the proximal region). • ETS factors, ELF4 and ELF1, and Pax5 and c-Myb bind and activate FBXO7 luciferase reporter constructs. • Fbxo7 represses transcription from its own promoter, and this is a ubiquitin ligase-independent effect. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
25. c-Myb interferes with inflammatory IL1α-NF-κB pathway in breast cancer cells.
- Author
-
Dúcka, Monika, Kučeríková, Martina, Trčka, Filip, Červinka, Jakub, Biglieri, Elisabetta, Šmarda, Jan, Borsig, Lubor, Beneš, Petr, and Knopfová, Lucia
- Subjects
- *
BREAST cancer , *CANCER cells , *RECOMBINANT proteins , *GENES , *CELL lines - Abstract
• c-Myb attenuates NF-κB activity in breast cancer. • Interaction with co-activator p300 is required for NF-κB suppression by c-Myb. • c-Myb negatively regulates IL1a transcription in several ER- breast cancer cell lines. • Inhibition of IL1α expression mediates the anti-inflammatory effect of c-Myb. The transcription factor c-Myb can be involved in the activation of many genes with protumorigenic function; however, its role in breast cancer (BC) development is still under discussion. c-Myb is considered as a tumor-promoting factor in the early phases of BC, on the other hand, its expression in BC patients relates to a good prognosis. Previously, we have shown that c-Myb controls the capacity of BC cells to form spontaneous lung metastasis. Reduced seeding of BC cells to the lungs is linked to high expression of c-Myb and a decline in expression of a specific set of inflammatory genes. Here, we unraveled a c-Myb-IL1α-NF-κB signaling axis that takes place in tumor cells. We report that an overexpression of c-Myb interfered with the activity of NF-κB in several BC cell lines. We identified IL1α to be essential for this interference since it was abrogated in the IL1α-deficient cells. Overexpression of IL1α, as well as addition of recombinant IL1α protein, activated NF-κB signaling and restored expression of the inflammatory signature genes suppressed by c-Myb. The endogenous levels of c-Myb negatively correlated with IL1α on both transcriptional and protein levels across BC cell lines. We concluded that inhibition of IL1α expression by c-Myb reduces NF-κB activity and disconnects the inflammatory circuit, a potentially targetable mechanism to mimic the antimetastatic effect of c-Myb with therapeutic perspective. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
26. c-myb is involved in CML progression and is a therapeutic target in the zebrafish CML model.
- Author
-
Ye Y, Yang X, Li F, Liu W, Zhang W, and Huang Z
- Subjects
- Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Fusion Proteins, bcr-abl genetics, Fusion Proteins, bcr-abl metabolism, Neutrophils drug effects, Neutrophils metabolism, Piperidines pharmacology, Piperidines therapeutic use, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Animals, Genetically Modified, Disease Models, Animal, Disease Progression, Flavonoids pharmacology, Flavonoids therapeutic use, Imatinib Mesylate pharmacology, Imatinib Mesylate therapeutic use, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Proto-Oncogene Proteins c-myb genetics, Proto-Oncogene Proteins c-myb metabolism, Zebrafish
- Abstract
Background: Despite the success of tyrosine kinase inhibitors in chronic myeloid leukemia (CML) therapy, CML still faces the challenges of drug resistance and progression to blast crisis. Twenty-five percent of patients have imatinib resistance and treatment difficulties due to heterogeneity after progression, but little is known about the mechanism. A key transcription factor in hematopoiesis, MYB, has been reported to increase abnormally in several types of aggressive blood disorders including CML., Methods: This study used a zebrafish model to explore the relationship between BCR/ABL1 and c-myb in CML progression. A CML zebrafish model was crossed with a c-myb hyperactivity transgenic line., Results: It was found that both exogenous BCR/ABL1 and c-myb could up-regulate the expression of neutrophil-related genes. More seriously, neutrophil accumulation was observed when BCR/ABL1 was combined with c-myb overexpression. Further studies showed that c-myb may be one of the downstream targets of BCR/ABL1 and the effect of BCR/ABL1 on neutrophils was c-myb dependent. Taking advantage of this inheritable in vivo model, it was shown that a combination of imatinib and flavopiridol, a cyclin-dependent kinase inhibitor targeting MYB, could more effectively alleviate the aggressive phenotype of the double transgene line., Conclusion: In summary, this study suggests that c-myb acts downstream of BCR/ABL1 and is involved in CML progression and is therefore a risk factor and a valuable target for the treatment of CML progression. The model used in the study could be helpful in high-throughput drug screening in CML transformation., (© 2022 The Authors. Animal Models and Experimental Medicine published by John Wiley & Sons Australia, Ltd on behalf of The Chinese Association for Laboratory Animal Sciences.)
- Published
- 2024
- Full Text
- View/download PDF
27. Semaphorin 5A drives melanoma progression: role of Bcl-2, miR-204 and c-Myb
- Author
-
Simona D’Aguanno, Elisabetta Valentini, Maria Grazia Tupone, Marianna Desideri, Marta Di Martile, Manuela Spagnuolo, Simonetta Buglioni, Cristiana Ercolani, Italia Falcone, Marco De Dominici, Michele Milella, Maria Giulia Rizzo, Bruno Calabretta, Carlo Cota, Andrea Anichini, Daniela Trisciuoglio, and Donatella Del Bufalo
- Subjects
Melanoma ,Semaphorin 5A ,Bcl-2 ,c-Myb ,miR-204 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Melanoma, the most aggressive form of skin cancer, is characterized by high rates of metastasis, drug resistance and mortality. Here we investigated the role of Semaphorin 5A (Sema5A) on the properties associated with melanoma progression and the factors involved in Sema5A regulation. Methods Western blotting, qRT-PCR, Chromatin immunoprecipitation (ChIP) assay, immunohistochemistry of melanoma patient specimens and xenograft tissues, in vitro Transwell assay for cell migration and invasion evaluation, in vitro capillary-like structure formation analysis. Results A significant correlation of Sema5A mRNA expression and melanoma progression was observed by analyzing GEO profile dataset. Endogenous Sema5A protein was detected in 95% of human melanoma cell lines tested, in 70% of metastatic specimens from patients affected by melanoma, and 16% of in situ melanoma specimens showed a focal positivity. We demonstrated that Sema5A regulates in vitro cell migration and invasion and the formation of vasculogenic structures. We also found an increase of Sema5A at both mRNA and protein level after forced expression of Bcl-2. By use of transcriptional and proteasome inhibitors, we showed that Bcl-2 increases the stability of Sema5A mRNA and protein. Moreover, by ChIP we demonstrated that Sema5A expression is under the control of the transcription factor c-Myb and that c-Myb recruitment on Sema5A promoter is increased after Bcl-2 overexpression. Finally, a concomitant decrease in the expression of Sema5A, Bcl-2 and c-Myb proteins was observed in melanoma cells after miR-204 overexpression. Conclusion Overall our data provide evidences supporting the role of Sema5A in melanoma progression and the involvement of Bcl-2, miR-204 and c-Myb in regulating its expression.
- Published
- 2018
- Full Text
- View/download PDF
28. Acute liver injury upregulates microRNA-491-5p in mice, and its overexpression sensitizes Hep G2 cells for tumour necrosis factor-alpha-induced apoptosis.
- Author
-
Wu, Jian and Zern, M A
- Subjects
Hepatology ,C-myb ,fibrosis ,hepatic stellate cells ,miRNA-150 ,miRNA-194 ,rac1 - Abstract
BACKGROUND: MicroRNAs (miRNAs) have emerged as novel genetic regulators of cell functions such as proliferation, apoptosis and cancer.AIMS: The aim of this study was to evaluate the role of a specific miRNA in modulating hepatic cell functions. Methods: C57Bl/6 mice were administered anti-fas receptor antibodies to induce liver cell apoptosis. miRNAs were purified from the liver tissue and evaluated using an miRNA microarray. The role of miRNA-491_5p, which was overexpressed in the model, in modulating hepatic cell functions was evaluated. miRNA-491_5p was overexpressed in Hep G2 cells, followed by the addition of tumour necrosis factor (TNF)-alpha, and induction of apoptosis as well as genes involved in apoptosis pathways were evaluated. The effect of miRNA-491_5p target genes on apoptosis was also analysed by inhibiting their expression by siRNA-induced gene silencing.RESULTS: Upregulation of miRNA-491_5p was found in a high-dose anti-fas receptor antibody group. Overexpression of microRNA-491_5p sensitized Hep G2 cells for TNF-alpha-induced apoptosis, and also caused an inhibition of alpha-fetoprotein, (AFP), heat shock protein-90 (hsp-90) and nuclear factor-kappaB (NF-kappaB). Overexpression of miRNA-491_5p or inhibition of AFP and hsp-90 resulted in an increased apoptosis in TNF-alpha-treated Hep G2 cells.CONCLUSIONS: One of the miRNAs that is associated with the acute liver injury mouse model, miRNA-491_5p, sensitizes Hep G2 cells for TNF-alpha-induced apoptosis, at least in part, by inhibiting AFP, hsp-90 and NF-kappaB.
- Published
- 2010
29. Liver fibrosis causes downregulation of miRNA-150 and miRNA-194 in hepatic stellate cells, and their overexpression causes decreased stellate cell activation.
- Author
-
Wu, Jian and Zern, M A
- Subjects
Hepatology ,C-myb ,fibrosis ,hepatic stellate cells ,miRNA-150 ,miRNA-194 ,rac1 - Abstract
Activation of hepatic stellate cells (HSC) results in their proliferation and in the secretion of extracellular matrix (ECM) proteins, which leads to hepatic fibrosis. microRNAs (miRNAs) have been shown to regulate various cell functions, such as proliferation, differentiation, and apoptosis. Hence, we have analyzed the miRNAs that were differentially expressed in HSC isolated from sham-operated and bile duct-ligated rats. Expression of two miRNAs, miRNA-150 and miRNA-194, was reduced in HSC isolated from fibrotic rats compared with sham-operated animals. These two miRNAs were overexpressed in LX-2 cells, and their ability to inhibit cell proliferation, the expression of smooth muscle alpha-actin (SMA), a marker for activation, and collagen type I, a marker for ECM secretion, was determined. Overexpression of these two miRNAs resulted in a significant inhibition of proliferation (P < 0.05) and reduced SMA and collagen I levels compared with either untreated cells or nonspecific miRNA-expressing cells. Next, the protein targets of these two miRNAs were found using bioinformatics approaches. C-myb was found to be a target for miRNA-150, and rac 1 was found to be one of the targets for miRNA-194. Therefore, we studied the expression of these two proteins by overexpressing these two miRNAs in LX-2 cells and found that overexpression of miRNA-150 and miRNA-194 resulted in a significant inhibition of c-myb and rac 1 expression, respectively. We conclude that both miRNA-150 and miRNA-194 inhibit HSC activation and ECM production, at least in part, via inhibition of c-myb and rac 1 expression.
- Published
- 2010
30. Dissecting the transactivation domain (tAD) of the transcription factor c‐Myb to assess recent models of tAD function.
- Author
-
Næs, Guro, Storesund, Jan Ove, Udayakumar, Priyanga‐Dina, Ledsaak, Marit, and Gabrielsen, Odd Stokke
- Subjects
TRANSCRIPTION factors ,PHASE separation ,GENETIC regulation ,TEST systems ,ALANINE - Abstract
Transcription factors use a DNA‐binding domain to localize their action and a transactivation domain (tAD) to stimulate activation of the associated gene. Recent work has renewed interest in how tADs activate genes, which remains poorly understood. Key features in the new models are exposure of short linear motifs (SLMs) and liquid–liquid phase separation (LLPS). Inspired by the new models for tAD function, we decided to revisit the tAD of the haematopoietic transcription factor c‐Myb by performing a mutational analysis to see how these new models fit and potentially explain the tAD behaviour of this master regulator. We know that c‐Myb has an acidic tAD, which contains a well‐characterized SLM in the form of a LxxLL motif. By testing 12 alanine‐scanning mutants and three mutants with major reorganization of its tAD in two mammalian reporter systems, we found a pattern of effects very close to what would be expected from the SLM‐exposure model, with strong effects exerted by both acidic replacements and SLM mutation. When the same mutants were tested in a yeast system, the pattern of effects was dramatically different, with the SLM mutation exerting no effect, and tAD behaviour was much less affected by small alterations, as would be expected from a LLPS model. These observations are discussed in the light of the two new tAD models, and a two‐step hypothesis for transactivation, combining both models, is proposed. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
31. Components from the Human c-myb Transcriptional Regulation System Reactivate Epigenetically Repressed Transgenes.
- Author
-
Barrett, Cassandra M., McCracken, Reilly, Elmer, Jacob, and Haynes, Karmella A.
- Subjects
- *
CHROMOSOMAL proteins , *TRANSGENES , *CHIMERIC proteins , *BINDING sites , *DNA , *PROMOTERS (Genetics) , *TRANSGENE expression - Abstract
A persistent challenge for mammalian cell engineering is the undesirable epigenetic silencing of transgenes. Foreign DNA can be incorporated into closed chromatin before and after it has been integrated into a host cell’s genome. To identify elements that mitigate epigenetic silencing, we tested components from the c-myb and NF-kB transcriptional regulation systems in transiently transfected DNA and at chromosomally integrated transgenes in PC-3 and HEK 293 cells. DNA binding sites for MYB (c-myb) placed upstream of a minimal promoter enhanced expression from transiently transfected plasmid DNA. We targeted p65 and MYB fusion proteins to a chromosomal transgene, UAS-Tk-luciferase, that was silenced by ectopic Polycomb chromatin complexes. Transient expression of Gal4-MYB induced an activated state that resisted complete re-silencing. We used custom guide RNAs and dCas9-MYB to target MYB to different positions relative to the promoter and observed that transgene activation within ectopic Polycomb chromatin required proximity of dCas9-MYB to the transcriptional start site. Our report demonstrates the use of MYB in the context of the CRISPR-activation system, showing that DNA elements and fusion proteins derived from c-myb can mitigate epigenetic silencing to improve transgene expression in engineered cell lines. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
32. MAGI1 mediates tumor metastasis through c-Myb/miR-520h/MAGI1 signaling pathway in renal cell carcinoma.
- Author
-
Wang, Wei, Yang, Yanhua, Chen, Xinyi, Shao, Shihong, Hu, Shasha, and Zhang, Tingguo
- Subjects
RENAL cell carcinoma ,METASTASIS ,ADHERENS junctions - Abstract
Renal cell carcinoma (RCC) is the third most common urological cancer with highly metastatic potential. MAGI1 plays an important role in stabilization of the adherens junctions and has been confirmed to suppress invasiveness and metastasis in multiple cancers in clinic. However, its expression and anti-metastatic ability in RCC are still unclear. In this study, we demonstrated that MAGI1 was markedly decreased in the RCC and indicated poor survival. Furthermore, we found that MAGI1 suppressed the invasion and migration of human RCC cells. Mechanistic investigations revealed that MAGI1 stabilized the PTEN/MAGI1/β-catenin complex to inhibit β-catenin signaling pathway. Moreover, MAGI1 was targeted by miR-520h which was transcriptionally activated by c-Myb. Collectively, our findings suggested that MAGI1mediated tumor metastasis through c-Myb/miR-520h/MAGI1 signaling pathway in RCC. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
33. miR-150 Regulates Memory CD8 T Cell Differentiation via c-Myb
- Author
-
Zeyu Chen, Erietta Stelekati, Makoto Kurachi, Sixiang Yu, Zhangying Cai, Sasikanth Manne, Omar Khan, Xiaolu Yang, and E. John Wherry
- Subjects
CD8 T cell ,T cell memory ,viral infection ,miR-150 ,c-Myb ,Bcl-2 ,Bcl-xl ,immune memory ,T cell differentiation ,Biology (General) ,QH301-705.5 - Abstract
MicroRNAs play an important role in T cell responses. However, how microRNAs regulate CD8 T cell memory remains poorly defined. Here, we found that miR-150 negatively regulates CD8 T cell memory in vivo. Genetic deletion of miR-150 disrupted the balance between memory precursor and terminal effector CD8 T cells following acute viral infection. Moreover, miR-150-deficient memory CD8 T cells were more protective upon rechallenge. A key circuit whereby miR-150 repressed memory CD8 T cell development through the transcription factor c-Myb was identified. Without miR-150, c-Myb was upregulated and anti-apoptotic targets of c-Myb, such as Bcl-2 and Bcl-xL, were also increased, suggesting a miR-150-c-Myb survival circuit during memory CD8 T cell development. Indeed, overexpression of non-repressible c-Myb rescued the memory CD8 T cell defects caused by overexpression of miR-150. Overall, these results identify a key role for miR-150 in memory CD8 T cells through a c-Myb-controlled enhanced survival circuit.
- Published
- 2017
- Full Text
- View/download PDF
34. Establishment of a zebrafish hematological disease model induced by 1,4-benzoquinone
- Author
-
Ao Zhang, Mei Wu, Junliang Tan, Ning Yu, Mengchang Xu, Xutong Yu, Wei Liu, and Yiyue Zhang
- Subjects
1,4-Benzoquinone ,Hematotoxicity ,Neutrophilia ,c-myb ,Zebrafish ,Medicine ,Pathology ,RB1-214 - Abstract
Benzene exposure is associated with various hematological disorders, in particular leukemia. The reactive metabolite of benzene, 1,4-benzoquinone (BQ), generated in bone marrow, is suggested to be a key molecule in mediating benzene-induced hematotoxicity and carcinogenicity. However, its pathogenic role remains largely unknown due to a lack of suitable vertebrate whole-organism models. Here, we present an in vivo study to reveal the effect of BQ exposure on hematotoxicity in zebrafish. From embryonic stages to adulthood, BQ exposure suppressed erythroid and lymphoid hematopoiesis but led to abnormal accumulation of myeloid cells and precursors, which resembles benzene-induced cytopenia and myeloid dysplasia in humans. This myeloid expansion is caused by granulocyte, but not macrophage, lineage, emphasizing the significant role of lineage specificity in BQ-mediated hematopoietic toxicity. Analysis of the c-myb (also known as myb)-deficient mutant cmybhkz3 revealed that BQ induced neutrophilia in a c-myb-dependent manner, demonstrating that c-myb is a key intrinsic mediator of BQ hematotoxicity. Our study reveals that BQ causes lineage-specific hematotoxicity in zebrafish from embryonic stages to adulthood. Since c-myb is indispensable for BQ to induce neutrophilia, c-myb could serve as a potential drug target for reversing BQ hematotoxicity.
- Published
- 2019
- Full Text
- View/download PDF
35. Modulation of Myb‐induced NF‐kB ‐STAT3 signaling and resulting cisplatin resistance in ovarian cancer by dietary factors.
- Author
-
Tian, Miao, Tian, Dan, Qiao, Xiaofang, Li, Jinlong, and Zhang, Leilei
- Subjects
- *
CISPLATIN , *EPIGALLOCATECHIN gallate , *OVARIAN cancer , *DRUG resistance in cancer cells , *TUMOR grading , *CANCER invasiveness - Abstract
c‐Myb regulates tumorigenesis in multiple cancers. Here we show, for the first time, the mechanism of c‐Myb‐mediated proliferation, invasion, and drug resistance in ovarian cancer (OC), the most lethal gynecological cancer, and a comparative analyses of dietary agents, curcumin, epigallocatechin‐3‐gallate (EGCG), and sulforaphane in inhibiting c‐Myb activity. We evaluated myb expression in patients with OC and found its increased expression in patients with cancer, compared with normal controls and in higher grade tumors, compared with low‐grade tumors. Using ES2 and OVCAR3 cell line models, along with the silencing or overexpression of c‐Myb, we establish a role of c‐Myb in determining resistance to cisplatin. c‐Myb overexpression activated NF‐κB and STAT3 signaling leading to enhanced proliferation, invasion, and cisplatin resistance. Contrary to this, silencing of c‐Myb inhibited proliferation, invasion, and sensitized OC cells to cisplatin. Further, among the dietary agents tested, EGCG almost completely inhibited the c‐Myb‐induced proliferation and invasion whereas sulforaphane also had significant inhibitory effect. Both compounds significantly sensitized OC cells to cisplatin, reversing the c‐Myb effects. Higher c‐Myb levels in patients with ovarian cancer lead to poor survival and our results indicate a possible effect of dietary factors EGCG and sulforaphane against c‐Myb‐mediated ovarian cancer progression and chemoresistance. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
36. c‐Myb promotes growth and metastasis of colorectal cancer through c‐fos‐induced epithelial‐mesenchymal transition.
- Author
-
Qu, Xiao, Yan, Xuebing, Kong, Cheng, Zhu, Yin, Li, Hao, Pan, Dengdeng, Zhang, Xiaohui, Liu, Yongqiang, Yin, Fang, and Qin, Huanlong
- Abstract
c‐Myb is a crucial transcription factor that participates in various biological functions; however, its role in colorectal cancer (CRC) remains poorly investigated. We first analyzed the expression and clinical significance of c‐Myb in a retrospective cohort enrolling 132 CRC patients. Then, the CRISPR/Cas9 technique was used to establish c‐Myb gene KO CRC cell lines. Cellular functional assays in vitro and in vivo were used to evaluate the impact of c‐Myb KO in CRC cells. Finally, RNA sequencing was used to investigate the potential oncogenic mechanisms regulated by c‐Myb in CRC progression and related cellular validations were accordingly carried out. As a result, c‐Myb is significantly overexpressed in CRC tissues as compared with adjacent normal tissues. High expression of c‐Myb is positively correlated with lymph node metastasis and poor prognosis. Univariate analysis and multivariate analysis further identify c‐Myb as an independent unfavorable prognostic factor for CRC patients. c‐Myb KO inhibits the proliferation, apoptosis resistance, invasion, metastasis, colony formation and in vivo tumorigenesis of CRC cells. Also, the mechanism investigation indicates that c‐Myb may promote CRC progression by regulating c‐fos. c‐fos overexpression can rescue the inhibitory effect of c‐Myb KO on the malignant characteristics of CRC cells. Finally, we find that c‐Myb KO inhibits the epithelial‐mesenchymal transition (EMT) molecular phenotype in CRC cells, whereas c‐fos overexpression can rescue this inhibitory effect. This study suggests that c‐Myb promotes the malignant progression of CRC through c‐fos‐induced EMT and has the potential to be a promising prognostic biomarker and therapeutic target. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
37. c‐MYB‐ and PGC1a‐dependent metabolic switch induced by MYBBP1A loss in renal cancer.
- Author
-
Felipe‐Abrio, Blanca, Verdugo‐Sivianes, Eva M., and Carnero, Amancio
- Abstract
The tumor microenvironment may alter the original tumorigenic potential of tumor cells. Under harsh environmental conditions, genetic alterations conferring selective advantages may initiate the growth of tumor subclones, providing new opportunities for these tumors to grow. We performed a genetic loss‐of‐function screen to identify genetic alterations able to promote tumor cell growth in the absence of glucose. We identified that downregulation of MYBBP1A increases tumorigenic properties under nonpermissive conditions. MYBBP1A downregulation simultaneously activates PGC1α, directly by alleviating direct repression and indirectly by increasing PGC1α mRNA levels through c‐MYB, leading to a metabolic switch from glycolysis to OXPHOS and increased tumorigenesis in low‐glucose microenvironments. We have also identified reduced MYBBP1A expression in human renal tumor samples, which show high expression levels of genes involved in oxidative metabolism. In summary, our data support the role of MYBBP1A as a tumor suppressor by regulating c‐MYB and PGC1α. Therefore, loss of MYBBP1A increases adaptability spanning of tumors through metabolic switch. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
38. MicroRNA‐1258, regulated by c‐Myb, inhibits growth and epithelial‐to‐mesenchymal transition phenotype via targeting SP1 in oral squamous cell carcinoma.
- Author
-
Zhang, Hua, Jiang, Sui, Guo, Longbin, and Li, Xi
- Subjects
SQUAMOUS cell carcinoma ,CELL growth ,CANCER invasiveness ,CELL lines ,WESTERN immunoblotting - Abstract
The biological function and underlying mechanism of miR‐1258 has seldom been investigated in cancer progression, including in oral squamous cell carcinoma (OSCC). In the current study, we revealed that the expression level of miR‐1258 was significantly down‐regulated in OSCC tissues and cell lines. Restoration of miR‐1258 decreased OSCC cell growth and invasion. The luciferase and Western blot assays revealed that SP1 protein was a downstream target of miR‐1258. Overexpression of SP1 dismissed miR‐1258's effect on cell growth and invasion. We also revealed that c‐Myb inhibited miR‐1258 by directly binding at its promoter. In addition, miR‐1258 inhibited PI3K/AKT and ERK signalling pathway activity. Taken together, these findings demonstrated that miR‐1258 may function as a tumour‐suppressive micorRNA in OSCC and suggested that miR‐1258 may be a potential therapeutic target for OSCC patients. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
39. B-Cell Deficiency Lowers Blood Pressure in Mice.
- Author
-
Dingwell, Luke S., Shikatani, Eric A., Besla, Rickvinder, Levy, Andrew S., Dinh, Danny D., Momen, Abdul, Zhang, Hangjun, Afroze, Talat, Chen, Michelle B., Chiu, Felix, Simmons, Craig A., Billia, Filio, Gommerman, Jennifer L., John, Rohan, Heximer, Scott, Scholey, James W., Bolz, Steffen-Sebastian, Robbins, Clinton S., and Husain, Mansoor
- Abstract
The proto-oncogene c-myb (and corresponding nuclear transcription factor, c-Myb) regulates the proliferation and differentiation of hematologic and vascular smooth muscle cells; however, the role of c-Myb in blood pressure regulation is unknown. Here, we show that mice homozygous for a hypomorphic c-myb allele ( c-myb h/h) conferring reduced c-Myb activity manifest reduced peripheral blood and kidney B220+ B-cells and have decreased systolic (104±2 versus 120±1 mm Hg; P<0.0001) and diastolic blood pressure (71±2 versus 83±1 mm Hg; P<0.0001) compared with WT (wild type) mice. Additionally, c-myb h/h mice had lower susceptibility to deoxycorticosterone acetate-salt experimental hypertension. Although cardiac (echocardiography) and resistance artery (perfusion myography) functions were normal, metabolic cage studies revealed that c-myb h/h mice had increased 24-hour urine output and sodium excretion versus WT. Reconstitution of WT mice with c-myb h/h bone marrow transplant and chimeric bone marrow transplant using mice lacking B-cells ( J H T; h/h>WT and h/h:J H T>WT, respectively) decreased blood pressure and increased 24-hour urine output compared with controls ( WT>WT; WT:J H T>WT). J H T mice also had decreased systolic (103±2 versus 115±1 mm Hg; P<0.0001) and diastolic blood pressure (71±2 versus 79±1; P<0.01) and increased 24-hour urine output versus WT. Real-time quantitative reverse transcription polymerase chain reaction of kidney medulla revealed reduced V2R (vasopressin receptor 2) expression in c-myb h/h and J H T mice. These data implicate B-cells in the regulation of V2R and its associated effects on salt and water handling and blood pressure homeostasis. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
40. c-MYB and DMTF1 in Cancer.
- Author
-
Fry, Elizabeth A. and Inoue, Kazushi
- Subjects
- *
TUMOR treatment , *CELL proliferation , *APOPTOSIS , *BREAST tumors , *CELL differentiation , *CELLULAR signal transduction , *GENE expression , *GENES , *LEUKEMIA , *NUCLEIC acid probes , *ONCOGENES , *SURVIVAL , *TRANSCRIPTION factors - Abstract
The c-Myb gene encodes a transcription factor that regulates cell proliferation, differentiation, and apoptosis through protein-protein interaction and transcriptional regulation of signaling pathways. The protein is frequently overexpressed in human leukemias, breast cancers, and other solid tumors suggesting that it is a bona fide oncogene. c-MYB is often overexpressed by translocation in human tumors with t(6;7)(q23;q34) resulting in c-MYB-TCRβ in T cell ALL, t(X;6)(p11;q23) with c-MYB-GATA1 in acute basophilic leukemia, and t(6;9)(q22-23;p23-24) with c-MYB-NF1B in adenoid cystic carcinoma. Antisense oligonucleotides to c-MYB were developed to purge bone marrow cells to eliminate tumor cells in leukemias. Recently, small molecules that inhibit c-MYB activity have been developed to disrupt its interaction with p300. The Dmp1 (cyclin D binding myb-like protein 1; Dmtf1) gene was isolated through its virtue for binding to cyclin D2. It is a transcription factor that has a Myb-like repeat for DNA binding. The Dmtf1 protein directly binds to the Arf promoter for transactivation and physically interacts with p53 to activate the p53 pathway. The gene is hemizygously deleted in 35-42% of human cancers and is associated with longer survival. The significances of aberrant expression of c-MYB and DMTF1 proteins in human cancers and their clinical significances are discussed. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
41. UM-HACC-2A: MYB-NFIB fusion-positive human adenoid cystic carcinoma cell line.
- Author
-
Warner, Kristy A., Oklejas, Alexandra E., Pearson, Alexander T., Zhang, Zhaocheng, Wu, Weishing, Divi, Vasu, Rodriguez-Ramirez, Christie, Castilho, Rogerio M., Polverini, Peter J., and Nör, Jacques E.
- Subjects
- *
ADENOID cystic carcinoma , *SUBMANDIBULAR gland , *CELL lines , *SALIVARY glands , *CELL suspensions - Abstract
Objectives: Limited availability of validated human adenoid cystic carcinoma (ACC) cell lines has hindered the mechanistic understanding of the pathobiology of this malignancy and the development of effective therapies. The purpose of this work was to generate and characterize a human ACC cell line.Material and Methods: Immediately after surgery, a tumor fragment from a minor salivary gland from the tongue of a female Caucasian was minced, dissociated, and a single cell suspension was plated in fibronectin-coated flasks. A culture medium containing bovine brain extract and rhEGF was optimized for these cells. Whole exome sequencing was used to evaluate the presence of MYB-NFIB translocation.Results: The University of Michigan-Human Adenoid Cystic Carcinoma (UM-HACC)-2A cells showed continuous growth in monolayers for at least 180 in vitro passages while maintaining epithelial morphology. Short-tandem repeat (STR) profiling confirmed a 100% match to patient DNA. Whole exome sequencing revealed the presence of the MYB-NFIB fusion in UM-HACC-2A cells, which was confirmed by PCR analysis. Western blots revealed high expression of epithelial markers (e.g. E-cadherin, EGFR, pan-cytokeratin) and proteins associated with ACC (e.g. c-Myb, p63). Developmental therapeutic studies showed that UM-HACC-2A cells were resistant to cisplatin (IC50 = 44.7 µM) while more responsive to paclitaxel (IC50 = 0.0006 µM). In a pilot study, we observed that UM-HACC-2A cells survived orthotopic transplantation into the submandibular gland. Notably, one of the mice injected with UM-HACC-2A cells exhibited lung metastasis after 6 months.Conclusion: UM-HACC-2A is a MYB-NFIB fusion-positive ACC cell line that is suitable for mechanistic and developmental therapeutics studies. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
42. Resveratrol Activates Natural Killer Cells through Akt- and mTORC2-Mediated c-Myb Upregulation
- Author
-
Yoo-Jin Lee and Jongsun Kim
- Subjects
NK cells ,resveratrol ,Akt ,mTOR complex 2 ,c-Myb ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Natural killer (NK) cells are suitable targets for cancer immunotherapy owing to their potent cytotoxic activity. To maximize the therapeutic efficacy of cancer immunotherapy, adjuvants need to be identified. Resveratrol is a well-studied polyphenol with various potential health benefits, including antitumor effects. We previously found that resveratrol is an NK cell booster, suggesting that it can serve as an adjuvant for cancer immunotherapy. However, the molecular mechanism underlying the activation of NK cells by resveratrol remains unclear. The present study aimed to determine this mechanism. To this end, we investigated relevant pathways in NK cells using Western blot, real-time polymerase chain reaction, pathway inhibitor, protein/DNA array, and cytotoxicity analyses. We confirmed the synergistic effects of resveratrol and interleukin (IL)-2 on enhancing the cytolytic activity of NK cells. Resveratrol activated Akt by regulating Mammalian Target of Rapamycin (mTOR) Complex 2 (mTORC2) via phosphatase and tensin homolog (PTEN) and ribosomal protein S6 kinase beta-1 (S6K1). Moreover, resveratrol-mediated NK cell activation was more dependent on the mTOR pathway than the Akt pathway. Importantly, resveratrol increased the expression of c-Myb, a downstream transcription factor of Akt and mTORC2. Moreover, c-Myb was essential for resveratrol-induced NK cell activation in combination with IL-2. Our results demonstrate that resveratrol activates NK cells through Akt- and mTORC2-mediated c-Myb upregulation.
- Published
- 2020
- Full Text
- View/download PDF
43. Effect of ischemic preconditioning on the expression of c-myb in the CA1 region of the gerbil hippocampus after ischemia/reperfusion injury
- Author
-
Hui Young Lee, Hyun-Jin Tae, Geum-Sil Cho, In Hye Kim, Jeong Hwi Cho, Joon Ha Park, Ji Hyeon Ahn, Bai Hui Chen, Bich-Na Shin, Moo-Ho Won, Chan Woo Park, Jun Hwi Cho, Jeong Yeol Seo, and Jae-Chul Lee
- Subjects
c-myb ,Cells in stratum pyramidale ,Delayed neuronal death ,Ischemic preconditioning ,Ischemia-reperfusion ,Protection ,Medicine - Abstract
Objective(s): In the present study, we investigated the effect of ischemic preconditioning (IPC) on c-myb immunoreactivity as well as neuronal damage/death after a subsequent lethal transient ischemia in gerbils. Materials and Methods: IPC was subjected to a 2 min sublethal ischemia and a lethal transient ischemia was given 5 min transient ischemia. The animals in all of the groups were given recovery times of 1 day, 2 days and 5 days and we examined change in c-myb immunoreactivity as well as neuronal damage/death in the hippocampus induced by a lethal transient ischemia. Results:A lethal transient ischemia induced a significant loss of cells in the stratum pyramidale (SP) of the hippocampal CA1 region at 5 days post-ischemia, and this insult showed that c-myb immunoreactivity in cells of the SP of the CA1 region was significantly decreased at 2 days post-ischemia and disappeared at 5 days post-ischemia. However, IPC effectively prevented the neuronal loss in the SP and showed that c-myb immunoreactivity was constitutively maintained in the SP after a lethal transient ischemia. Conclusion: Our results show that a lethal transient ischemia significantly decreased c-myb immunoreactivity in the SP of the CA1 region and that IPC well preserved c-myb immunoreactivity in the SP of the CA1 region. We suggest that the maintenance of c-myb might be related with IPC-mediated neuroprotection after a lethal ischemic insult.
- Published
- 2016
44. MicroRNA-150 Inhibits the Activation of Cardiac Fibroblasts by Regulating c-Myb
- Author
-
Peng Deng, Ling Chen, Zheng Liu, Ping Ye, Sihua Wang, Jie Wu, Yufeng Yao, Yuan Sun, Xiaofan Huang, Linyun Ren, Anchen Zhang, Ke Wang, Chuangyan Wu, Zhang Yue, Xuezeng Xu, and Manhua Chen
- Subjects
MiR-150 ,Cardiac fibrosis ,Cardiac fibroblast ,Myofibroblast ,c-Myb ,Physiology ,QP1-981 ,Biochemistry ,QD415-436 - Abstract
Background/Aims: Cardiac fibrosis is the primary cause of deteriorated cardiac function in various cardiovascular diseases. Numerous studies have demonstrated that microRNAs (miRNAs) are critical regulators of myocardial fibrosis. Specifically, many studies have reported that miR-150 is downregulated in cardiovascular diseases, such as acute myocardial infarction (AMI), myocardial hypertrophy and myocardial fibrosis. However, the exact role of miR-150 in these pathological processes remains unknown. Methods: We used the transverse aortic constriction (TAC) mouse model to study the role of miR-150 in cardiac fibrosis induced by pressure overload. After the TAC operation, qRT-PCR was used to measure the expression profiles of miR-150 in left ventricle tissues and populations of primary heart cell types. Then, we used both miR-150 knockout mice and wild type (WT) mice in the TAC model. Changes in cardiac function and pathology were measured using transthoracic echocardiography and pathological analysis, respectively. Furthermore, we predicted the target of miR-150 in cardiac fibroblasts (CFs) and completed in vitro CF transfection experiments using miR-150 analogs and siRNA corresponding to the predicted target. Results: We observed decreased expression levels of miR-150 in hearts suffering pressure overload, and these levels decreased more sharply in CFs than in cardiomyocytes. In addition, the degrees of cardiac function deterioration and cardiac fibrosis in miR-150-/- mice were more severe than were those in WT mice. By transfecting CFs with an miR-150 analog in vitro, we observed that miR-150 inhibited cardiac fibroblast activation. We predicted that the transcription factor c-Myb was the target of miR-150 in CFs. Transfecting CFs with c-Myb siRNA eliminated the effects of an miR-150 inhibitor, which promoted CF activation. Conclusion: These findings reveal that miR-150 acts as a pivotal regulator of pressure overload-induced cardiac fibrosis by regulating c-Myb.
- Published
- 2016
- Full Text
- View/download PDF
45. Components from the Human c-myb Transcriptional Regulation System Reactivate Epigenetically Repressed Transgenes
- Author
-
Cassandra M. Barrett, Reilly McCracken, Jacob Elmer, and Karmella A. Haynes
- Subjects
myb ,c-myb ,transgene ,epigenetic silencing ,activator ,heterochromatin ,polycomb ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
A persistent challenge for mammalian cell engineering is the undesirable epigenetic silencing of transgenes. Foreign DNA can be incorporated into closed chromatin before and after it has been integrated into a host cell’s genome. To identify elements that mitigate epigenetic silencing, we tested components from the c-myb and NF-kB transcriptional regulation systems in transiently transfected DNA and at chromosomally integrated transgenes in PC-3 and HEK 293 cells. DNA binding sites for MYB (c-myb) placed upstream of a minimal promoter enhanced expression from transiently transfected plasmid DNA. We targeted p65 and MYB fusion proteins to a chromosomal transgene, UAS-Tk-luciferase, that was silenced by ectopic Polycomb chromatin complexes. Transient expression of Gal4-MYB induced an activated state that resisted complete re-silencing. We used custom guide RNAs and dCas9-MYB to target MYB to different positions relative to the promoter and observed that transgene activation within ectopic Polycomb chromatin required proximity of dCas9-MYB to the transcriptional start site. Our report demonstrates the use of MYB in the context of the CRISPR-activation system, showing that DNA elements and fusion proteins derived from c-myb can mitigate epigenetic silencing to improve transgene expression in engineered cell lines.
- Published
- 2020
- Full Text
- View/download PDF
46. Human MicroRNA Targetome Indicates a Specialized Role of MicroRNAs in Regulation of Oncogenesis
- Author
-
Satoh, Jun-ichi and Azmi, Asfar S., editor
- Published
- 2012
- Full Text
- View/download PDF
47. Utilization of circular dichroism and electrospray ionization mass spectrometry to understand the formation and conversion of G-quadruplex DNA at the human c-myb proto-oncogene.
- Author
-
Fu, Hengqing, Yang, Pengfei, Hai, Jinhui, and Li, Huihui
- Subjects
- *
CIRCULAR dichroism , *OPTICAL polarization , *ONCOGENES , *DNA , *IONIZING radiation - Abstract
G-quadruplex DNAs are involved in a number of key biological processes, including gene expression, transcription, and apoptosis. The c-myb oncogene contains a number of GGA repeats in its promoter which forms G-quadruplex, thus it could be used as a target in cancer therapeutics. Several in-vitro studies have used Circular Dichroism (CD) spectroscopy or electrospray ionization mass spectrometry (ESI-MS) to demonstrate formation and stability of G-quadruplex DNA structure in the promoter region of human c-myb oncogene. The factors affecting the c-myb G-quadruplex structures were investigated, such as cations (i.e. K + , NH 4 + and Na + ) and co-solutes (methanol and polyethylene glycol). The results indicated that the presence of cations and co-solutes could change the G-quadruplex structural population and promote its thermodynamic stabilization as indicated by CD melting curves. It indicated that the co-solutes preferentially stabilize the c-myb G-quadruplex structure containing both homo- and hetero-stacking. In addition, protopine was demonstrated as a binder of c-myb G-quadruplex as screened from a library of natural alkaloids using ESI-MS method. CD spectra showed that it could selectively stabilize the c-myb G-quadruplex structure compared to other six G-quadruplexes from tumor-related G-rich sequences and the duplex DNAs (both long and short-chain ones). The binding of protopine could induce the change in the G-quadruplex structural populations. Therefore, protopine with its high binding specificity could be considered as a precursor for the design of drugs to target and regulate c-myb oncogene transcription. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
48. Role of c-Myb in the regulation of natural killer cell activity.
- Author
-
Shin, Hee-Wook, Lee, Yoo-Jin, and Kim, Jongsun
- Subjects
- *
IMMUNOTHERAPY , *TRANSCRIPTION factors - Abstract
Abstract The regulation of natural killer (NK) cell activity is an important research goal for the development of immunotherapies. In this study, we identified transcription factors affecting NK cell activity. In particular, we screened transcription factors affected by interleukin-2 (IL-2) and transforming growth factor-beta (TGF-β) by protein/DNA arrays using primary NK cells. We found that celastrol, a c-Myb inhibitor, inhibited NK-92 cells more strongly than any other inhibitors of transcription factor candidates. In addition, c-Myb and c-Myb-related signaling molecules, e.g., Nemo-like kinase (NLK) and c-Myc, were regulated by the activation status of NK cells, suggesting that c-Myb is a key regulator of NK cell activity. We also found that celastrol inhibits NK-92-cell-mediated cytotoxicity via the downregulation of NKG2D and granzyme B. Knockdown studies also showed that c-Myb is important for NK cell activation. In particular, the knockdown of c-Myb did not significantly affect NK cell proliferation and survival but decreased the secretion of IFN-γ and the cytotoxicity of NK cells. Our data demonstrate that c-Myb plays a critical role in the activation of NK cells and therefore is a therapeutic target for cancer and viral diseases. Graphical abstract Image 1 Highlights • c-Myb activity was downregulated by TGF-β in natural killer cells. • Celastrol, a c-Myb inhibitor, decreased NK-92 activity at low concentrations. • Celastrol downregulated NKG2D and granzyme B expression. • c-Myb knockdown downregulated NK-92 cytotoxicity and IFN-γ secretion. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
49. Low expression of Mda-7/IL-24 and high expression of C-myb in tumour tissues are predictors of poor prognosis for Burkitt lymphoma patients.
- Author
-
Ma, Ming, Zhao, Riyang, Yang, Xingxiao, Zhao, Lianmei, Liu, Lihua, Zhang, Cong, Wang, Xuexiao, and Shan, Baoen
- Subjects
- *
BURKITT'S lymphoma , *MYB gene , *PROGNOSIS , *IMMUNOHISTOCHEMISTRY , *LYMPH nodes , *PATIENTS - Abstract
Objectives: Burkitt lymphoma is one of the most common types of haematopoietic malignancy in children and adolescents. Mda-7/IL-24 had been identified as a differentiation inducer of Burkitt lymphoma cells. Previous studies have revealed that knockdown of C-myb can also lead to the terminal differentiation of Burkitt lymphoma cells. The aim of the present study was to investigate the correlation between the expression of Mda-7/IL-24 and C-myb, as well as their prognostic significance, for Burkitt lymphoma patients. Methods: The tumour tissues were collected from 59 cases of Burkitt lymphoma patients and detected with Western blotting and immunohistochemistry. Results: The results showed that the expression of Mda-7/IL-24 was lower, whereas the expression of C-myb was higher in Burkitt lymphoma tissues compared to specimens of normal lymph node tissues. Furthermore, C-myb expression was negatively correlated with Mda-7/IL-24 expression at the protein level in Burkitt lymphoma tissues and cell lines. Both the expression of Mda-7/IL-24 and C-myb in Burkitt lymphoma tissues was associated with some clinicopathological parameters, such as clinical stage, infiltration in the bone marrow, Ki67 and overall survival rates. Conclusion: These results indicated that low expression of Mda-7/IL-24 along with high expression of C-myb are predictors for poor prognosis of Burkitt lymphoma patients; this outcome suggests that Mda-7/IL-24 and C-myb might be potential targets for clinical treatment of Burkitt lymphoma. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
50. The clinical significance of Mda-7/IL-24 and C-myb expression in tumor tissues of patients with diffuse large B cell lymphoma.
- Author
-
Ma, Ming, Zhao, Riyang, Yang, Xingxiao, Zhao, Lianmei, Liu, Lihua, Zhang, Cong, Wang, Xuexiao, and Shan, Baoen
- Subjects
- *
DIFFUSE large B-cell lymphomas , *CYTOKINES , *LYMPHOMAS , *GENE expression , *PHENOTYPES , *THERAPEUTICS , *PROGNOSIS - Abstract
Diffuse large B cell lymphoma (DLBCL) is the most common type of non-Hodgkin's lymphoma in adults. Mda-7/IL-24 had been identified as a differentiation inducer of B phenotype lymphoma cells. Previous studies have revealed that knockdown of C-myb also leads to the terminal differentiation of B cell lymphoma. The aim of the present study was to investigate the association between the expression of Mda-7/IL-24 and C-myb, and their prognostic significance for DLBCL patients. The tumor tissues were collected from 72 cases of DLBCL patients and detected with reverse transcription-quantitative polymerase chain reaction, western blotting and immunohistochemistry assays. The results showed that, the expression of Mda-7/IL-24 mRNA and protein was lower while the expression of C-myb was higher in DLBCL tissues, compared with the specimens of normal lymph node tissues. Furthermore, C-myb expression was negatively correlated with Mda-7/IL-24 expression at mRNA and protein levels in DLBCL tissues. The expression of Mda-7/IL-24 and C-myb in DLBCL tissues was associated with some clinicopathological parameters such as clinical stage, infiltration in bone marrow, Ki67 expression level in the tumor tissues and overall survival rates. These results indicated that low expression of Mda-7/IL-24, along with high expression of C-myb, are predictor for poor prognosis of DLBCL patients, suggesting that Mda-7/IL-24 and C-myb may be potential targets for clinical treatment of DLBCL. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.