1
|
Leung CCT, Wong CKC. Characterization of stanniocalcin-1 expression in macrophage differentiation. Transl Oncol 2020; 14:100881. [PMID: 33074126 PMCID: PMC7568195 DOI: 10.1016/j.tranon.2020.100881] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/30/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023] Open
Abstract
Human stanniocalcin-1 (STC1) is a paracrine factor associated with inflammation and carcinogenesis. The role of STC1 in the pro- and anti-inflammatory functions of differentiating macrophage, however, is not clear. In this study, our data showed that phorbol 12-myristate 13-acetate (PMA) treatment induced human leukemia monocytic cells (ThP-1) differentiation to M0 macrophages. The differentiation was accompanied by a significant increase in the mRNA expression levels of STC1, the pro-inflammatory cytokine TNFα, and anti-inflammatory markers, CD163 & CD206. An intermitted removal of PMA treatment reduced the mRNA levels of STC1 and TNFα but had no noticeable effects on the anti-inflammatory markers. The correlation in the expression of STC1 and pro-inflammatory markers in differentiating macrophages was investigated, using siRNASTC1-transfected PMA-induced cells. Consistently, the transcripts levels of TNFα and IL-6 were significantly reduced. Moreover, LPS/IFNγ-induced M1-polarization showed remarkably higher expression levels of STC1 than IL-4/IL-13-induced M2-macrophages and PMA-induced M0-macrophages. Transcriptomic analysis of siRNASTC1-transfected M1-polarized cells revealed an upregulation of TBC1 domain family member 3 (TBC1D3G). The gene regulates the payload of macrophage-released extracellular vesicles to mediate inflammation. The conditioned media from siRNASTC1-transfected M1-polarized cells were found to reduce Hep3B cell motility. The data suggest that the expression of STC1 were associated with macrophage differentiation, but preferentially to M1 polarization.
Collapse
Affiliation(s)
- Cherry C T Leung
- Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China
| | - Chris K C Wong
- Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
2
|
Tang W, Gao H, Li J, Wang X, Zhou Z, Gai L, Feng XJ, Tian J, Lu H, Guo Z. A General Strategy for the Construction of NIR-emitting Si-rhodamines and Their Application for Mitochondrial Temperature Visualization. Chem Asian J 2020; 15:2724-2730. [PMID: 32666700 DOI: 10.1002/asia.202000660] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/07/2020] [Indexed: 11/07/2022]
Abstract
Si-rhodamine (SiR) is an ideal fluorophore because it possesses bright emission in the NIR region and can be implemented flexibly in living cells. Currently, several promising approaches for synthesizing SiR are being developed. However, challenges remain in the construction of SiR containing functional groups for bioimaging application. Herein, we introduce a general and simple approach by a condensation reaction of diarylsilylether and arylaldehyde in o-dichlorobenzene to synthesize a series of SiRs bearing various functional substituents. These SiRs have moderate to high quantum efficiency, tolerance to photobleaching, and high water solubility as well as NIR emitting, and their NIR fluorescence properties can be controlled through the photoinduced electron transfer (PET) mechanism. Fluorescence OFF-ON switching effect is observed for SiR 9 in the presence of acid, which is rationalized by DFT/TDDFT calculations. Moreover, reversible stimuli response toward temperature is achieved. Since positive charge enables mitochondrial targeting ability and chloromethyl unit can covalently immobilize the dyes onto the mitochondrial via click reaction between the benzyl choride and protein sulfhydryls, SiR 8 is identified as a valuable fluorescent marker to visualize the morphology and monitor the temperature change of mitochondria with high photostability.
Collapse
Affiliation(s)
- Weiguo Tang
- Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education and Key Laboratory of Organosilicon Material of Zhejiang Province, Hangzhou Normal University, No. 2318, Yuhangtang Road, Hangzhou, 311121, P. R. China
| | - Han Gao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Jiaxin Li
- Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education and Key Laboratory of Organosilicon Material of Zhejiang Province, Hangzhou Normal University, No. 2318, Yuhangtang Road, Hangzhou, 311121, P. R. China
| | - Xianhui Wang
- Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education and Key Laboratory of Organosilicon Material of Zhejiang Province, Hangzhou Normal University, No. 2318, Yuhangtang Road, Hangzhou, 311121, P. R. China
| | - Zhikuan Zhou
- Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education and Key Laboratory of Organosilicon Material of Zhejiang Province, Hangzhou Normal University, No. 2318, Yuhangtang Road, Hangzhou, 311121, P. R. China
| | - Lizhi Gai
- Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education and Key Laboratory of Organosilicon Material of Zhejiang Province, Hangzhou Normal University, No. 2318, Yuhangtang Road, Hangzhou, 311121, P. R. China
| | - Xin Jiang Feng
- Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education and Key Laboratory of Organosilicon Material of Zhejiang Province, Hangzhou Normal University, No. 2318, Yuhangtang Road, Hangzhou, 311121, P. R. China
| | - Jiangwei Tian
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Hua Lu
- Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education and Key Laboratory of Organosilicon Material of Zhejiang Province, Hangzhou Normal University, No. 2318, Yuhangtang Road, Hangzhou, 311121, P. R. China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
3
|
Bhattacharya A, Ghosh P, Prasad R, Ghosh A, Das K, Roy A, Mallik S, Sinha DK, Sen P. MAP Kinase driven actomyosin rearrangement is a crucial regulator of monocyte to macrophage differentiation. Cell Signal 2020; 73:109691. [PMID: 32531262 DOI: 10.1016/j.cellsig.2020.109691] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 12/31/2022]
Abstract
Rearrangement of actin cytoskeleton correlates significantly with the immune responses as the perturbation of cytoskeletal dynamics leads to many immune deficiencies. Mechanistic insights into this correlation remain unknown. Cellular spreading, the most characteristic phenotype associated with monocyte to macrophage differentiation, led us to investigate the contribution of actomyosin dynamics in monocyte differentiation. Our observation revealed that actomyosin reorganization intrinsically governs the process of monocyte to macrophage differentiation. Further, we established that the MAPK-driven signaling pathways regulate the cellular actomyosin dynamics that direct monocyte to macrophage differentiation. We also identified P42/44 Mitogen-Activated Protein Kinase (P42/44 MAPK), P38 Mitogen-Activated Protein Kinase (P38 MAPK), MAP Kinase Activated Protein Kinase 2 (MK-2), Heat Shock Protein 27 (Hsp-27), Lim Kinase (Lim K), non-muscle cofilin (n-cofilin), Myosin Light Chain Kinase (MLCK) and Myosin Light Chain (MLC) as critical components of the signaling network. Moreover, we have shown the involvement of the same signaling cascade in 3D gel-like microenvironment induced spontaneous monocyte to macrophage differentiation and in human blood-derived PBMC differentiation. Our study reveals new mechanistic insights into the process of monocyte to macrophage differentiation.
Collapse
Affiliation(s)
- Anindita Bhattacharya
- Department of Biological Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Purnam Ghosh
- Department of Biological Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Ramesh Prasad
- Department of Biological Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Arnab Ghosh
- Department of Biological Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Kaushik Das
- Department of Biological Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Abhishek Roy
- Department of Biological Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Suman Mallik
- Department of Biological Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Deepak Kumar Sinha
- Department of Biological Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Prosenjit Sen
- Department of Biological Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India.
| |
Collapse
|
4
|
Fixable Molecular Thermometer for Real-Time Visualization and Quantification of Mitochondrial Temperature. Anal Chem 2018; 90:13953-13959. [PMID: 30422634 DOI: 10.1021/acs.analchem.8b03395] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A change of mitochondrial temperature can be an important indicator of mitochondrial metabolism that generates considerable heat. For this reason, development of fluorescent probes to detect mitochondrial temperature has become an attractive topic. Previous efforts have successfully addressed the major issues, such as temperature sensitivity and mitochondrial targetability. However, there remains a key obstacle to practical applications. Considering the highly dynamic features of mitochondria, especially the variation of the inner-membrane potential, it is quite necessary to permanently immobilize a temperature probe in mitochondria in order to avoid unstable intracellular localization along with the changes of mitochondrial status. Herein, we report Mito-TEM, the first fixable, fluorescent molecular thermometer. Mito-TEM is based on a positively charged rhodamine B fluorophore that has the tendency of being attracted to mitochondria, which have negative potential. This fluorophore containing rotatable substituents also contributes to the temperature-responsive fluorescence property. Most importantly, a benzaldehyde is introduced in Mito-TEM as an anchoring unit that condenses with aminos of the protein and thus immobilizes the probe in mitochondria. The specific immobilization of Mito-TEM in mitochondria is unambiguously demonstrated in colocalization imaging. By using Mito-TEM, a method of visualizing and quantifying a temperature distribution through grayscale imaging of mitochondria is established and further applied to monitor the temperature changes of live cells under light heating and PMA stimulation.
Collapse
|
5
|
Muiwo P, Pandey P, Ahmad HM, Ramachandran SS, Bhattacharya A. IsomiR processing during differentiation of myelogenous leukemic cell line K562 by phorbol ester PMA. Gene 2017; 641:172-179. [PMID: 29051025 DOI: 10.1016/j.gene.2017.10.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 05/18/2017] [Accepted: 10/10/2017] [Indexed: 01/28/2023]
Abstract
Chronic myelocytic leukemia cell line K562 undergoes differentiation by phorbol esters to megakaryocytes and we have used this system to understand miRNA processing leading to isomiR generation. PMA treatment significantly altered the production of miRNA in K562 cells. Expression of 24.4% of miRNAs were found to be stimulated whereas expression of 10% miRNAs were inhibited by PMA treatment. Our results suggest that miRNA precursors are processed into isomiRs in a deterministic manner. The relative levels of different isomiRs of a miRNA remained mainly unchanged even after PMA treatment irrespective of overall changes in expression (either up-regulation or down-regulation). However, not all miRNAs behave in the same way, about 7% showed a variation of isomiR profiles after PMA treatment. Most of the later class of miRNAs were found to be oncogenic miRNAs. Further, it was also found that number of isomiRs was independent of abundance of a miRNA. Functional importance of different isomiRs was demonstrated using three different isomiRs of miR-22. Our results showed that different isomiRs could inhibit expression of targets genes with different efficiencies. Our study suggests that the heterogeneity of a miRNA population generated during processing is in general regulated and that variation in the generation of an isomiR can be a functionally important regulatory feature.
Collapse
Affiliation(s)
- Pamchui Muiwo
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.
| | - Priyatama Pandey
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, India.
| | - Hafiz M Ahmad
- Department of Molecular Cell and Cancer Biology, Umass Medical School, Worcester, MA, USA.
| | | | - Alok Bhattacharya
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India; School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
6
|
miR-125b modulates megakaryocyte maturation by targeting the cell-cycle inhibitor p19 INK4D. Cell Death Dis 2016; 7:e2430. [PMID: 27763644 PMCID: PMC5133966 DOI: 10.1038/cddis.2016.288] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 08/16/2016] [Accepted: 08/17/2016] [Indexed: 12/16/2022]
Abstract
A better understanding of the mechanisms involved in megakaryocyte maturation will facilitate the generation of platelets in vitro and their clinical applications. A microRNA, miR-125b, has been suggested to have important roles in the self-renewal of megakaryocyte-erythroid progenitors and in platelet generation. However, miR-125b is also critical for hematopoietic stem cell self-renewal. Thus, the function of miR-125b and the complex signaling pathways regulating megakaryopoiesis remain to be elucidated. In this study, an attentive examination of the endogenous expression of miR-125b during megakaryocyte differentiation was performed. Accordingly, the differentiation of hematopoietic stem cells requires the downregulation of miR-125b, whereas megakaryocyte determination and maturation synchronize with miR-125b accumulation. The overexpression of miR-125b improves megakaryocytic differentiation of K562 and UT-7 cells. Furthermore, stage-specific overexpression of miR-125b in primary cells demonstrates that miR-125b mediates an enhancement of megakaryocytic differentiation after megakaryocyte determination, the stage at which megakaryocytes are negative for the expression of the hematopoietic progenitor marker CD34. The identification of miR-125b targets during megakaryopoiesis was focused on negative regulators of cell cycle because the transition of the G1/S phase has been associated with megakaryocyte polyploidization. Real-time PCR, western blot and luciferase reporter assay reveal that p19INK4D is a direct target of miR-125b. P19INK4D knockdown using small interfering RNA (siRNA) in megakaryocyte-induced K562 cells, UT-7 cells and CD61+ promegakaryocytes results in S-phase progression and increased polyploidy, as well as improved megakaryocyte differentiation, similarly to the effects of miR-125b overexpression. P19INK4D overexpression reverses these effects, as indicated by reduced expression of megakaryocyte markers, G1-phase arrest and polyploidy decrease. P19INK4D knockdown in miR-125b downregulated cells or p19INK4D overexpression in miR-125b upregulated cells rescued the effect of miR-125b. Taken together, these findings suggest that miR-125b expression positively regulates megakaryocyte development since the initial phases of megakaryocyte determination, and p19INK4D is one of the key mediators of miR-125b activity during the onset of megakaryocyte polyploidization.
Collapse
|
7
|
Ho YH, Yao CL, Lin KH, Hou FH, Chen WM, Chiang CL, Lin YN, Li MW, Lin SH, Yang YJ, Lin CC, Lu J, Tigyi G, Lee H. Opposing regulation of megakaryopoiesis by LPA receptors 2 and 3 in K562 human erythroleukemia cells. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:172-83. [PMID: 25463482 DOI: 10.1016/j.bbalip.2014.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 11/06/2014] [Accepted: 11/13/2014] [Indexed: 01/10/2023]
Abstract
Erythrocytes and megakaryocytes (MK) are derived from a common progenitor that undergoes lineage specification. Lysophosphatidic acid (LPA), a lipid growth factor was previously shown to be a regulator for erythropoietic process through activating LPA receptor 3 (LPA3). However, whether LPA affects megakaryopoiesis remains unclear. In this study, we used K562 leukemia cell line as a model to investigate the roles of LPA in MK differentiation. We demonstrated that K562 cells express both LPA2 and LPA3, and the expression levels of LPA2 are higher than LPA3. Treatment with phorbol 12-myristate 13-acetate, a commonly used inducer of megakaryopoiesis, reciprocally regulates the expressions of LPA2 and LPA3. By pharmacological blockers and knockdown experiments, we showed that activation of LPA2 suppresses whereas, LPA3 promotes megakaryocytic differentiation in K562. The LPA2-mediated inhibition is dependent on β-catenin translocation, whereas reactive oxygen species (ROS) generation is a downstream signal for activation of LPA3. Furthermore, the hematopoietic transcriptional factors GATA-1 and FLI-1, appear to be involved in these regulatory mechanisms. Taken together, our results suggested that LPA2 and LPA3 may function as a molecular switch and play opposing roles during megakaryopoiesis of K562 cells.
Collapse
Affiliation(s)
- Ya-Hsuan Ho
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Chao-Ling Yao
- Department of Chemical Engineering and Materials Science, Yuan-Ze University, Chung-Li, Taiwan, ROC
| | - Kuan-Hung Lin
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Fen-Han Hou
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Wei-Min Chen
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Chi-Ling Chiang
- School of Biomedical Science, The Ohio State University, Columbus, USA
| | - Yu-Nung Lin
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Meng-Wei Li
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Shi-Hung Lin
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Ya-Jan Yang
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Chu-Cheng Lin
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Jenher Lu
- Department of Pediatrics and Pediatric Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC.
| | - Gabor Tigyi
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, USA.
| | - Hsinyu Lee
- Department of Life Science, National Taiwan University, Taipei, Taiwan, ROC; Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan, ROC; Angiogenesis Research Center, National Taiwan University, Taipei, Taiwan, ROC; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan, ROC; Center for Biotechnology, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
8
|
Ahmad HM, Muiwo P, Ramachandran SS, Pandey P, Gupta YK, Kumar L, Kulshreshtha R, Bhattacharya A. miR-22 regulates expression of oncogenic neuro-epithelial transforming gene 1, NET1. FEBS J 2014; 281:3904-19. [DOI: 10.1111/febs.12926] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 05/17/2014] [Accepted: 07/08/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Hafiz M. Ahmad
- School of Life Sciences; Jawaharlal Nehru University; New Delhi India
| | - Pamchui Muiwo
- School of Life Sciences; Jawaharlal Nehru University; New Delhi India
| | | | - Priyatama Pandey
- School of Computational and Integrative Sciences; Jawaharlal Nehru University; New Delhi India
| | - Yogendra K. Gupta
- Department of Pharmacology; All India Institute of Medical Sciences; New Delhi India
| | - Lalit Kumar
- Department of Medical Oncology; Institute Rotary Cancer Hospital; All India Institute of Medical Science; New Delhi India
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology; Indian Institute of Technology; New Delhi India
| | - Alok Bhattacharya
- School of Life Sciences; Jawaharlal Nehru University; New Delhi India
| |
Collapse
|
9
|
Huang R, Zhao L, Chen H, Yin RH, Li CY, Zhan YQ, Zhang JH, Ge CH, Yu M, Yang XM. Megakaryocytic differentiation of K562 cells induced by PMA reduced the activity of respiratory chain complex IV. PLoS One 2014; 9:e96246. [PMID: 24817082 PMCID: PMC4015910 DOI: 10.1371/journal.pone.0096246] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 04/06/2014] [Indexed: 01/14/2023] Open
Abstract
Mitochondria are involved in the regulation of cell differentiation processes, but its function changes and molecular mechanisms are not yet clear. In this study, we found that mitochondrial functions changed obviously when K562 cells were induced to megakaryocytic differentiation by phorbol 12-myristate 13-acetate (PMA). During the cell differentiation, the reactive oxygen species (ROS) level was increased, mitochondrial membrane potential declined and respiratory chain complex IV activity was decreased. Treatment with specific inhibitor of mitochondrial respiratory chain complex IV led to a significant inhibition in mitochondrial membrane potential and reduction of PMA-induced cell differentiation. However, treatment with cyclosporine A, a stabilization reagent of mitochondrial membrane potential, did not improve the down-regulation of mitochondrial respiratory chain complex IV induced by PMA. Furthermore, we found that the level of the complex IV core subunit COX3 and mitochondrial transport-related proteins Tim9 and Tim10 were decreased during the differentiation of K562 cells induced by PMA, suggesting an important role of these factors in mitochondrial functional changes. Our results suggest that changes in mitochondrial functions are involved in the PMA-induced K562 cell differentiation process, and the maintenance of the steady-state of mitochondrial functions plays a critical role in the regulation of cell differentiation.
Collapse
Affiliation(s)
- Rui Huang
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Long Zhao
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hui Chen
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Rong-Hua Yin
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Chang-Yan Li
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yi-Qun Zhan
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Jian-Hong Zhang
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Chang-hui Ge
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Miao Yu
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiao-Ming Yang
- Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
10
|
Lin HP, Chang JY, Lin SR, Lee MH, Huang SS, Hsu LJ, Chang NS. Identification of an In Vivo MEK/WOX1 Complex as a Master Switch for Apoptosis in T Cell Leukemia. Genes Cancer 2011; 2:550-62. [PMID: 21901168 DOI: 10.1177/1947601911418498] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 07/04/2011] [Indexed: 12/19/2022] Open
Abstract
Not all leukemia T cells are susceptible to high levels of phorbol myristate acetate (PMA)-mediated apoptosis. At micromolar levels, PMA induces apoptosis of Jurkat T cells by causing mitochondrial polarization/de-polarization, release of cytosolic granules, and DNA fragmentation. Chemical inhibitors U0126 and PD98059 block mitogen-activated protein kinase kinase 1 (MEK1)-mediated phosphorylation of extracellular signal-regulated kinase (ERK) and prevent apoptosis. Mechanistically, proapoptotic tumor suppressor WOX1 (also named WWOX or FOR) physically interacts with MEK1, in part, in the lysosomes in Jurkat cells. PMA induces the dissociation, which leads to relocation of MEK1 to lipid rafts and WOX1 to the mitochondria for causing apoptosis. U0126 inhibits PMA-induced dissociation of WOX1/MEK1 complex and supports survival of Jurkat cells. In contrast, less differentiated Molt-4 T cells are resistant to PMA-induced dissociation of the WOX1/MEK1 complex and thereby are refractory to apoptosis. U0126 overturns the resistance for enhancing apoptosis in Molt-4 cells. Together, the in vivo MEK1/WOX1 complex is a master on/off switch for apoptosis in leukemia T cells.
Collapse
Affiliation(s)
- Hsin-Ping Lin
- Institute of Molecular Medicine, National Cheng Kung University Medical College, Tainan, Taiwan
| | | | | | | | | | | | | |
Collapse
|
11
|
Sengupta S, Park SH, Seok GE, Patel A, Numata K, Lu CL, Kaplan DL. Quantifying osteogenic cell degradation of silk biomaterials. Biomacromolecules 2010; 11:3592-9. [PMID: 21105641 DOI: 10.1021/bm101054q] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The degradation of silk protein films by human mesenchymal stem cells (hMSCs), osteoblasts and osteoclasts, cells involved in osteogenic functions in normal and diseased bone, was assessed in vitro. The involvement of specific matrix metalloproteinases (MMPs) and integrin signaling in the degradation process was determined. Scanning electron microscopy (SEM) and atomic force microscopy (AFM) were used to quantitatively compare degradation by the different cell types using surface patterned silk films. Osteoblasts and osteoclasts demonstrated significant degradation of the silk films in vitro in comparison to the hMSCs and the film controls without cells. The osteoclasts degraded the silk films the most and also generated the highest level of MMPs 1 and 2. The osteoblasts upregulated integrins α5 and β1, while the osteoclasts upregulated integrins α2 and β1. There was significant contrast in responses on the silk matrices between osteogenic cells versus undifferentiated hMSCs to illustrate in vitro the role of cell type on matrix remodeling. These are important issues in matching biomaterial matrix features and studies in vitro to remodeling in vivo, in both normal and disease tissue systems. Cell populations and niche factors impact tissue regeneration, wound healing, physiological state, and the ability to better understand the role of different cell types is critical to overall regenerative outcomes.
Collapse
Affiliation(s)
- Sejuti Sengupta
- Biomedical Engineering, School of Engineering, Tufts University, 4 Colby Street, Medford, Massachussets 02155, United States
| | | | | | | | | | | | | |
Collapse
|
12
|
Lin YF, Lee HM, Leu SJ, Tsai YH. The essentiality of PKCα and PKCβI translocation for CD14+monocyte differentiation towards macrophages and dendritic cells, respectively. J Cell Biochem 2007; 102:429-41. [PMID: 17455194 DOI: 10.1002/jcb.21305] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Human peripheral CD14(+)monocytes have been known to differentiate into monocyte-derived macrophages (MDMs) or dendritic cells (MoDCs) upon suitable stimulation. However, the key intracellular molecule(s) associated with their differentiation towards specific cell types was(were) not fully understood. This study was designated to determine the association of PKC isoenzymes with the differentiation of CD14(+)monocytes into MDMs or MoDCs. Purified human peripheral CD14(+)monocytes were cultured with GM-CSF, or GM-CSF plus IL-4 for 7 days to induce cell differentiation. The phenotypic changes were analyzed by Flow-Cytometry using various specific antibodies to cell type-specific surface markers. The immunological functions of these differentiated cells were determined by measuring the amounts of TNF-alpha secretion for MDMs, and the capacities of antigen-capturing and bacterial phagocytosis for MoDCs. The translocations of PKC isoenzymes in these cells from cytosol to plasma membrane were examined by Western Blot analysis and Confocal Microscopic observation. The treatment of CD14(+)monocytes with either GM-CSF or PMA elicited PKCalpha translocation and consequently induced their differentiation into MDMs. The inclusion of PKCalpha/beta(I) specific inhibitor, Go6976, greatly inhibited the GM-CSF-induced PKCalpha translocation and dose-dependently reduced the GM-CSF- induced MDM differentiation. On the other hand, the simultaneous pretreatment of CD14(+)monocytes with Go6976 and PKCbeta-specific inhibitor predominantly suppressed the GM-CSF/IL-4-induced generation of MoDCs. Further study demonstrated that GM-CSF/IL-4 selectively induced the translocation of PKCbeta(I), not PKCalpha or PKCbeta(II), in CD14(+)monocytes. In conclusion, the cell fate commitment of CD14(+)monocytes towards MDMs or MoDCs appears to be steered by the selective activation of PKCalpha or PKCbeta(I), respectively.
Collapse
Affiliation(s)
- Yuan-Feng Lin
- Graduate Institute of Pharmaceutical Science, Taipei Medical University, Taipei, Taiwan
| | | | | | | |
Collapse
|
13
|
Arinze IJ, Kawai Y. Transcriptional Activation of the Human Gαi2 Gene Promoter through Nuclear Factor-κB and Antioxidant Response Elements. J Biol Chem 2005; 280:9786-95. [PMID: 15640523 DOI: 10.1074/jbc.m414006200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Very little is known regarding molecular mechanism(s) underlying transcriptional regulation of any G-protein gene despite the importance of G-protein expression in modulating cellular processes. Here we show that phorbol myristate acetate (PMA) and tert-butylhydroquinone (tBHQ), which induce oxidative stress in cells, up-regulate transcription of Galpha(i2) in K562 cells. Redox-sensing chemicals abrogated this transcriptional effect. A dominant negative I-kappaB double mutant (S32A/S36A) suppressed PMA-induced transcription by 54-62%, suggesting involvement of nuclear factor-kappaB (NF-kappaB). SN50, a cell-permeable peptide that inhibits nuclear import of stress-responsive transcription factors (such as NF-kappaB), inhibited PMA- and tBHQ-induced transcription. Deletion of an NF-kappaB-binding motif that maps at +10/+19 in the promoter resulted in 55-60% suppression of PMA-induced transcription, and 81% suppression of tBHQ-induced transcription. Mutation of an antioxidant response element (ARE) that maps at -84/-76 in the promoter resulted in 51 and 86% decrease in PMA- and tBHQ-induced transcription, respectively. In electrophoretic mobility shift assays, this element formed complexes with the transcription factors NF-E2p45 and Nrf2 that are prototypic for binding to the ARE, as well as with c-Fos, which can also interact with the ARE. Chromatin immunoprecipitation analysis demonstrated recruitment of these transcription factors to the promoter. Exogenously transfected Nrf2 transactivated the Galpha(i2) gene promoter; the cytoskeleton-associated protein, Keap1, abrogated this effect. Taken together, the present studies reveal that transcription factors that bind NF-kappaB and/or antioxidant response elements play an activating role in the transcription of the human Galpha(i2) gene.
Collapse
Affiliation(s)
- Ifeanyi J Arinze
- Department of Biochemistry, Meharry Medical College, 1005 David B. Todd Jr., Blvd, Nashville, Tennessee 37208-3599, USA.
| | | |
Collapse
|
14
|
Kim KW, Kim SH, Lee EY, Kim ND, Kang HS, Kim HD, Chung BS, Kang CD. Extracellular signal-regulated kinase/90-KDA ribosomal S6 kinase/nuclear factor-kappa B pathway mediates phorbol 12-myristate 13-acetate-induced megakaryocytic differentiation of K562 cells. J Biol Chem 2001; 276:13186-91. [PMID: 11278385 DOI: 10.1074/jbc.m008092200] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Two signaling pathways, the extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK)-dependent pathway and the nuclear factor-kappaB (NF-kappaB)-dependent pathway, have been known to mediate megakaryocytic differentiation of K562 cells induced by phorbol 12-myristate 13-acetate (PMA). In this study, we examined whether 90-kDa ribosomal S6 kinase (RSK), known as a substrate of ERK/MAPK and a signal-inducible IkappaBalpha kinase, would link two pathways during the differentiation. RSK1 was activated in a time- and dose-dependent manner during the PMA-induced differentiation. Overexpression of wild-type or dominant inhibitory mutant (D205N) of RSK1 enhanced or suppressed PMA-stimulated NF-kappaB activation and megakaryocytic differentiation as shown by morphology, nonspecific esterase activity, and expression of the CD41 megakaryocytic marker, respectively. In addition, overexpression of the dominant inhibitory mutant (S32A/S36A) of IkappaBalpha inhibited PMA-stimulated and RSK1-enhanced megakaryocytic differentiation, indicating that NF-kappaB mediates a signal for megakaryocytic differentiation downstream of RSK1. PMA-stimulated activation of ERK/MAPK, RSK1, and NF-kappaB and the PMA-induced megakaryocytic differentiation were prevented by pretreatment with PD98059, a specific inhibitor of the mitogen-activated ERK kinase (MEK). Therefore, these results demonstrate that the sequential ERK/RSK1/NF-kappaB pathway mediates PMA-stimulated megakaryocytic differentiation of K562 cells.
Collapse
Affiliation(s)
- K W Kim
- Department of Biochemistry, Research Center for Molecular Medicine, Pusan National University, Pusan 602-739, Korea
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Woessmann W, Mivechi NF. Role of ERK activation in growth and erythroid differentiation of K562 cells. Exp Cell Res 2001; 264:193-200. [PMID: 11262176 DOI: 10.1006/excr.2000.5124] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Inhibition of signaling through Ras in BCR-ABL-positive pluripotent K562 cells leads to apoptosis and spontaneous differentiation. However, Ras-induced activation of the mitogen-activated protein kinase ERK has been suggested to play a critical role in either growth or differentiation in different model systems. We studied the role of ERK activation in the growth-promoting and anti-apoptotic effect of Ras and its involvement in hemin-induced nonterminal erythroid differentiation using the BCR-ABL-positive K562 cell line as a model. K562 cells were stably transfected with ERK1 or the dominant inhibitory mutant of ERK1 (ERK1-KR). Overexpression of ERK1-KR inhibited cell growth with an approximately fourfold increase in doubling time and induced apoptosis in K562 cells. Incubation with the MEK1 inhibitor UO126 inhibited cell growth and induced apoptosis in K562 cells in a dose-dependent manner as well. In the presence of exogenously added hemin, K562 cells differentiate into erythroblasts, as indicated by the production of large amounts of fetal hemoglobin. We examined the activation of MAP kinases during hemin-induced differentiation. The ERK1 and 2 activity increased within 2 h post hemin treatment and remained elevated for 24-48 h. During this time, fetal hemoglobin synthesis also increases from 0.8 to 10 pg/cell. There was no activation of JNK or p38 protein kinases. The hemin-induced accumulation of hemoglobin was inhibited in ERK1-KR overexpressing cells and was enhanced in the wild-type ERK1 transfectants. Our results suggest that ERK activation is involved in both growth and hemin-induced erythroid differentiation in the BCR-ABL-positive K562 cell line.
Collapse
Affiliation(s)
- W Woessmann
- Institute of Molecular Medicine and Genetics, Department of Radiology, Medical College of Georgia, 1120 15th Street, Augusta, Georgia 30912, USA.
| | | |
Collapse
|
16
|
Hmama Z, Nandan D, Sly L, Knutson KL, Herrera-Velit P, Reiner NE. 1alpha,25-dihydroxyvitamin D(3)-induced myeloid cell differentiation is regulated by a vitamin D receptor-phosphatidylinositol 3-kinase signaling complex. J Exp Med 1999; 190:1583-94. [PMID: 10587349 PMCID: PMC2195730 DOI: 10.1084/jem.190.11.1583] [Citation(s) in RCA: 145] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
1alpha,25-dihydroxyvitamin D(3) (D(3)) promotes the maturation of myeloid cells and surface expressions of CD14 and CD11b, markers of cell differentiation in response to D(3). To examine how these responses are regulated, THP-1 cells were grown in serum-free medium and incubated with D(3). This was associated with rapid and transient increases in phosphatidylinositol 3-kinase (PI 3-kinase) activity. Furthermore, induction of CD14 expression in response to D(3) was abrogated by (a) the PI 3-kinase inhibitors LY294002 and wortmannin; (b) antisense oligonucleotides to mRNA for the p110 catalytic subunit of PI 3-kinase; and (c) a dominant negative mutant of PI 3-kinase. In THP-1 cells, induction of CD11b expression by D(3) was also abrogated by LY294002 and wortmannin. Similarly, LY294002 and wortmannin inhibited D(3)-induced expression of both CD14 and CD11b in peripheral blood monocytes. In contrast to CD14 and CD11b, hormone-induced expression of the Cdk inhibitor p21 in THP-1 cells was unaffected by either wortmannin or LY294002. These findings suggest that PI 3-kinase selectively regulates D(3)-induced monocyte differentiation, independent of any effects on p21.
Collapse
Affiliation(s)
- Zakaria Hmama
- Department of Medicine (Division of Infectious Diseases), The University of British Columbia, Faculties of Medicine and Science, The Research Institute of the Vancouver Hospital and Health Sciences Center, Vancouver, British Columbia, Canada V5Z 3J5
- Laboratoire d'Immunologie, Faculté des Sciences Dhar Mahraz, Université Mohamed Ben Abdallah, Fès, Morocco BP1796
| | - Devki Nandan
- Department of Medicine (Division of Infectious Diseases), The University of British Columbia, Faculties of Medicine and Science, The Research Institute of the Vancouver Hospital and Health Sciences Center, Vancouver, British Columbia, Canada V5Z 3J5
| | - Laura Sly
- Department of Medicine (Division of Infectious Diseases), The University of British Columbia, Faculties of Medicine and Science, The Research Institute of the Vancouver Hospital and Health Sciences Center, Vancouver, British Columbia, Canada V5Z 3J5
| | - Keith L. Knutson
- Department of Medicine (Division of Infectious Diseases), The University of British Columbia, Faculties of Medicine and Science, The Research Institute of the Vancouver Hospital and Health Sciences Center, Vancouver, British Columbia, Canada V5Z 3J5
| | - Patricia Herrera-Velit
- Department of Medicine (Division of Infectious Diseases), The University of British Columbia, Faculties of Medicine and Science, The Research Institute of the Vancouver Hospital and Health Sciences Center, Vancouver, British Columbia, Canada V5Z 3J5
| | - Neil E. Reiner
- Department of Medicine (Division of Infectious Diseases), The University of British Columbia, Faculties of Medicine and Science, The Research Institute of the Vancouver Hospital and Health Sciences Center, Vancouver, British Columbia, Canada V5Z 3J5
- Department of Microbiology and Immunology, The University of British Columbia, Faculties of Medicine and Science, The Research Institute of the Vancouver Hospital and Health Sciences Center, Vancouver, British Columbia, Canada V5Z 3J5
| |
Collapse
|
17
|
Kang CD, Han CS, Kim KW, Do IR, Kim CM, Kim SH, Lee EY, Chung BS. Activation of NF-kappaB mediates the PMA-induced differentiation of K562 cells. Cancer Lett 1998; 132:99-106. [PMID: 10397459 DOI: 10.1016/s0304-3835(98)00165-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The role of NF-kappaB during the PMA-induced megakaryocytic differentiation of K562 cells was investigated using K562 cells transfected with each or both subunits of NF-kappaB. The NF-kappaB subunit-transfected cells have shown much higher sensitivity to PMA-induced differentiation than their parental cells. This result was consistent with the findings that PMA-stimulated activities of NF-kappaB were markedly increased in the NF-kappaB subunit-transfected cells in comparison with their parental cells and PMA-induced differentiation was enhanced by pretreatment with IkappaB-alpha antisense oligonucleotide in the NF-kappaB subunit-transfected cells. Meanwhile, there were basically no difference in the basal and PMA-stimulated MAP kinase activities among the parental and NF-kappaB subunit-transfected cells, respectively. However, PMA-induced differentiation was blocked by pretreatment with PD98059, a specific inhibitor of MEK, in both parental and NF-kappaB-transfected cells. Therefore, these results suggest that during the PMA-induced megakaryocytic differentiation of K562 cells, NF-kappaB works downstream of MAP kinase, or that activation of both NF-kappaB and MAP kinase pathways is involved.
Collapse
Affiliation(s)
- C D Kang
- Pusan Cancer Research Center, Pusan National University, South Korea.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Ragg SJ, Kaga S, Berg KA, Ochi A. The Mitogen-Activated Protein Kinase Pathway Inhibits Ceramide-Induced Terminal Differentiation of a Human Monoblastic Leukemia Cell Line, U937. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.3.1390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
This communication describes an extracellular signal-regulated kinase kinase (MEK)-dependent signal transduction pathway that prevents the terminal differentiation of a hemopoietic cell line. Both PMA and the cell-permeable ceramide, C2-ceramide, caused differentiation of U937 cells, but with distinct cell morphology and CD11b/CD14 surface expression. While PMA activated extracellular signal-regulated kinase (ERK), a downstream kinase of Raf-MEK signaling, C2-ceramide activated c-Jun NH2-terminal kinase (JNK), an anchor kinase of stress-induced signaling. Furthermore, only C2-ceramide stimulated an induction of cell cycle arrest that was associated with stable expression of p21CIP1 and retinoblastoma nuclear phosphoprotein dephosphorylation. Expression of p21CIP1 and JNK activation were also observed in sphingosine-treated cells, whereas sphingosine did not induce detectable differentiation. Concomitant stimulation with C2-ceramide and PMA resulted in the PMA phenotype, and cell cycle arrest was absent. ERK activation was enhanced by C2-ceramide plus PMA stimulation, whereas the activation of JNK was aborted. Strikingly, the inhibition of MEK with PD98059 altered the phenotype of C2-ceramide- and PMA-stimulated U937 cells to that of cells treated with C2-ceramide alone. Thus, ERK and JNK pathways deliver distinct signals, and the ERK pathway is dominant to the JNK cascade. Furthermore, differentiation and cell cycle arrest caused by C2-ceramide rely on independent signaling pathways, and JNK is an unlikely signaling element for this differentiation. Importantly, during C2-ceramide and PMA costimulation, the JNK pathway is not simply blocked by ERK activation; rather, cross-talk between these MAP kinase pathways acts to simultaneously augment ERK activity and down-regulate JNK activity.
Collapse
Affiliation(s)
| | | | - Karen A. Berg
- †Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Atsuo Ochi
- *John P. Robarts Research Institute and
- †Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
19
|
Abstract
NF-κB/Rel designates a widely distributed family of transcription factors involved in immune and acute phase responses. Here, the expression and function of NF-κB factors in erythroid proliferation and differentiation were explored. In an erythroleukemia cell line, TF-1, high levels of p105/p50, p100/p52, p65, and IκBα were detected 24 hours after growth factor deprivation. In response to granulocyte-macrophage colony-stimulating factor (GM-CSF) stimulation, significant induction of p52 expression was observed. GM-CSF also induced nuclear translocation of both p52 and p65. No induction of NF-κB factors was observed with erythropoietin stimulation of TF-1 cells. Overexpression of p52 and p65 in TF-1 cells by transient transfection resulted in significant induction of a κB-TATA-luciferase reporter plasmid, showing that these factors are functional in vivo in erythroid cells. To determine whether NF-κB factors may play a role in normal erythropoiesis, levels of these factors were determined in burst-forming unit-erythroid (BFU-E)–derived cells at different stages of differentiation. The NF-κB factors p105/p50, p100/p52, and p65 were highly expressed in early BFU-E–derived precursors, which are rapidly proliferating, and declined during maturation. Furthermore, nuclear levels of NF-κB factors p50, p52, and p65 were higher in less mature precursors (day 10 BFU-E–derived cells) compared with more differentiated (day 14) erythroblasts. In nuclear extracts from day 10 BFU-E–derived cells, p50, p52, and p65 were able to form complexes, which bound to κB sites in the promoters of both the c-myb and c-mycgenes, suggesting that c-myb and c-myc may be among the κB-containing genes regulated by NF-κB factors in normal erythroid cells. Taken together, these data show that NF-κB factors are modulated by GM-CSF and suggest they function to regulate specific κB containing genes involved in erythropoiesis.
Collapse
|
20
|
Abstract
Abstract
NF-κB/Rel designates a widely distributed family of transcription factors involved in immune and acute phase responses. Here, the expression and function of NF-κB factors in erythroid proliferation and differentiation were explored. In an erythroleukemia cell line, TF-1, high levels of p105/p50, p100/p52, p65, and IκBα were detected 24 hours after growth factor deprivation. In response to granulocyte-macrophage colony-stimulating factor (GM-CSF) stimulation, significant induction of p52 expression was observed. GM-CSF also induced nuclear translocation of both p52 and p65. No induction of NF-κB factors was observed with erythropoietin stimulation of TF-1 cells. Overexpression of p52 and p65 in TF-1 cells by transient transfection resulted in significant induction of a κB-TATA-luciferase reporter plasmid, showing that these factors are functional in vivo in erythroid cells. To determine whether NF-κB factors may play a role in normal erythropoiesis, levels of these factors were determined in burst-forming unit-erythroid (BFU-E)–derived cells at different stages of differentiation. The NF-κB factors p105/p50, p100/p52, and p65 were highly expressed in early BFU-E–derived precursors, which are rapidly proliferating, and declined during maturation. Furthermore, nuclear levels of NF-κB factors p50, p52, and p65 were higher in less mature precursors (day 10 BFU-E–derived cells) compared with more differentiated (day 14) erythroblasts. In nuclear extracts from day 10 BFU-E–derived cells, p50, p52, and p65 were able to form complexes, which bound to κB sites in the promoters of both the c-myb and c-mycgenes, suggesting that c-myb and c-myc may be among the κB-containing genes regulated by NF-κB factors in normal erythroid cells. Taken together, these data show that NF-κB factors are modulated by GM-CSF and suggest they function to regulate specific κB containing genes involved in erythropoiesis.
Collapse
|
21
|
Herrera R, Hubbell S, Decker S, Petruzzelli L. A role for the MEK/MAPK pathway in PMA-induced cell cycle arrest: modulation of megakaryocytic differentiation of K562 cells. Exp Cell Res 1998; 238:407-14. [PMID: 9473349 DOI: 10.1006/excr.1997.3847] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In vitro megakaryocytic differentiation of the pluripotent K562 human leukemia cell line is induced by PMA. Treatment of K562 cells with PMA results in growth arrest, polyploidy, morphological changes, and increased cell-cell and cell-substrate adhesion. These PMA-induced changes in K562 cells are preceded by a rapid rise in the activity of MEK (MAP kinase/extracellular regulated kinases) that leads to a sustained activation of ERK2 (extracellular regulated kinase; MAPK). Blockade of MEK1 activation by PD098059, a recently described specific MEK inhibitor [D. T. Dudley et al. (1995). Proc. Natl. Acad. Sci. USA 92, 7686-7689], reverses both the growth arrest and the morphological changes of K562 cells induced by PMA treatment. These changes are not associated with a disruption of PMA-induced down-regulation of BCR-ABL kinase or early integrin signaling events but are associated with a block of the cell-surface expression of the gpIIb/IIIa (CD41) integrin, a cell marker of megakaryocytic differentiation. These results demonstrate that the PMA-induced signaling cascade initiated by protein kinase C activation requires the activity of the MEK/ERK signaling complex to regulate cell cycle arrest, thus regulating the program that leads to the cell-surface expression of markers associated with megakaryocytic differentiation.
Collapse
Affiliation(s)
- R Herrera
- Department of Cell Biology, Parke-Davis Pharmaceutical Research Division of Warner-Lambert Company, Ann Arbor, Michigan 48105, USA.
| | | | | | | |
Collapse
|