LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 25

Search options

  1. Article ; Online: DYRK2 controls a key regulatory network in chronic myeloid leukemia stem cells.

    Park, Chun Shik / Lacorazza, H Daniel

    Experimental & molecular medicine

    2020  Volume 52, Issue 10, Page(s) 1663–1672

    Abstract: Chronic myeloid leukemia is a hematological cancer driven by the oncoprotein BCR-ABL1, and lifelong treatment with tyrosine kinase inhibitors extends patient survival to nearly the life expectancy of the general population. Despite advances in the ... ...

    Abstract Chronic myeloid leukemia is a hematological cancer driven by the oncoprotein BCR-ABL1, and lifelong treatment with tyrosine kinase inhibitors extends patient survival to nearly the life expectancy of the general population. Despite advances in the development of more potent tyrosine kinase inhibitors to induce a durable deep molecular response, more than half of patients relapse upon treatment discontinuation. This clinical finding supports the paradigm that leukemia stem cells feed the neoplasm, resist tyrosine kinase inhibition, and reactivate upon drug withdrawal depending on the fitness of the patient's immune surveillance. This concept lends support to the idea that treatment-free remission is not achieved solely with tyrosine kinase inhibitors and that new molecular targets independent of BCR-ABL1 signaling are needed in order to develop adjuvant therapy to more efficiently eradicate the leukemia stem cell population responsible for chemoresistance and relapse. Future efforts must focus on the identification of new targets to support the discovery of potent and safe small molecules able to specifically eradicate the leukemic stem cell population. In this review, we briefly discuss molecular maintenance in leukemia stem cells in chronic myeloid leukemia and provide a more in-depth discussion of the dual-specificity kinase DYRK2, which has been identified as a novel actionable checkpoint in a critical leukemic network. DYRK2 controls the activation of p53 and proteasomal degradation of c-MYC, leading to impaired survival and self-renewal of leukemia stem cells; thus, pharmacological activation of DYRK2 as an adjuvant to standard therapy has the potential to induce treatment-free remission.
    MeSH term(s) Animals ; Carrier Proteins/metabolism ; Cell Self Renewal/genetics ; Disease Susceptibility ; Energy Metabolism ; Gene Expression Regulation, Leukemic ; Humans ; Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology ; Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism ; Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology ; Neoplastic Stem Cells/metabolism ; Neoplastic Stem Cells/pathology ; Protein Binding ; Protein Serine-Threonine Kinases/genetics ; Protein Serine-Threonine Kinases/metabolism ; Protein-Tyrosine Kinases/genetics ; Protein-Tyrosine Kinases/metabolism ; Signal Transduction ; Dyrk Kinases
    Chemical Substances Carrier Proteins ; Protein-Tyrosine Kinases (EC 2.7.10.1) ; Protein Serine-Threonine Kinases (EC 2.7.11.1)
    Language English
    Publishing date 2020-10-16
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 1328915-9
    ISSN 2092-6413 ; 1226-3613 ; 0378-8512
    ISSN (online) 2092-6413
    ISSN 1226-3613 ; 0378-8512
    DOI 10.1038/s12276-020-00515-5
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Retroviral Transduction of Quiescent Murine Hematopoietic Stem Cells.

    Park, Chun Shik / Lacorazza, H Daniel

    Methods in molecular biology (Clifton, N.J.)

    2017  Volume 1686, Page(s) 173–182

    Abstract: Hematopoietic stem cells (HSCs) represent an important target cell population in bone marrow transplantation, cell and gene therapy applications, and the development of leukemia models for research. Because the hematopoietic progeny carries the genetic ... ...

    Abstract Hematopoietic stem cells (HSCs) represent an important target cell population in bone marrow transplantation, cell and gene therapy applications, and the development of leukemia models for research. Because the hematopoietic progeny carries the genetic information of HSCs and replenishes the blood and immune system, corrective gene transfer into HSCs provides an ideal therapeutic approach for many monogenic hematological diseases and a useful tool for studies of HSC function and blood formation in normal and malignant hematopoiesis. However, the efficiency of gene transfer into HSCs has been limited by several features of viral vectors, viral titer, methods of viral transduction, and the property of stem cell quiescence. In this chapter, we describe the production of retrovirus using murine stem cell virus (MSCV)-based retroviral vectors and purification and transduction of murine HSCs.
    MeSH term(s) Animals ; Antigens, CD34/metabolism ; Cells, Cultured ; Flow Cytometry ; Genetic Vectors ; Hematopoietic Stem Cells/metabolism ; Mice ; Resting Phase, Cell Cycle ; Retroviridae/genetics ; Transduction, Genetic
    Chemical Substances Antigens, CD34
    Language English
    Publishing date 2017-10-13
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Research Support, N.I.H., Extramural
    ISSN 1940-6029
    ISSN (online) 1940-6029
    DOI 10.1007/978-1-4939-7371-2_13
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Role of the reprogramming factor KLF4 in blood formation.

    Park, Chun Shik / Shen, Ye / Lewis, Andrew / Lacorazza, H Daniel

    Journal of leukocyte biology

    2016  Volume 99, Issue 5, Page(s) 673–685

    Abstract: Krüppel-like factor 4 is a zinc finger protein with dual functions that can act as a transcriptional activator and repressor of genes involved in cell proliferation, differentiation, and apoptosis. Although most studies have focused on terminally ... ...

    Abstract Krüppel-like factor 4 is a zinc finger protein with dual functions that can act as a transcriptional activator and repressor of genes involved in cell proliferation, differentiation, and apoptosis. Although most studies have focused on terminally differentiated epithelial cells, evidence suggests that Krüppel-like factor 4 regulates the development and function of the myeloid and lymphoid blood lineages. The ability of Krüppel-like factor 4 to dedifferentiate from somatic cells into pluripotent stem cells in cooperation with other reprogramming factors suggests its potential function in the preservation of tissue-specific stem cells. Additionally, emerging interest in the redifferentiation of induced pluripotent stem cells into blood cells to correct hematologic deficiencies and malignancies warrants further studies on the role of Krüppel-like factor 4 in steady-state blood formation.
    MeSH term(s) Animals ; Cellular Reprogramming/genetics ; Chromatin/metabolism ; Gene Expression Regulation ; Hematopoiesis/genetics ; Humans ; Kruppel-Like Transcription Factors/genetics ; Kruppel-Like Transcription Factors/metabolism ; Models, Biological
    Chemical Substances Chromatin ; GKLF protein ; Kruppel-Like Transcription Factors
    Language English
    Publishing date 2016-05
    Publishing country United States
    Document type Journal Article ; Review
    ZDB-ID 605722-6
    ISSN 1938-3673 ; 0741-5400
    ISSN (online) 1938-3673
    ISSN 0741-5400
    DOI 10.1189/jlb.1RU1215-539R
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Stromal-induced epithelial-mesenchymal transition induces targetable drug resistance in acute lymphoblastic leukemia.

    Park, Chun Shik / Yoshihara, Hiroki / Gao, Qingsong / Qu, Chunxu / Iacobucci, Ilaria / Ghate, Pankaj S / Connelly, Jon P / Pruett-Miller, Shondra M / Wagner, Ben / Robinson, Camenzind G / Mishra, Ashutosh / Peng, Junmin / Yang, Lei / Rankovic, Zoran / Finkelstein, David / Luger, Selina / Litzow, Mark / Paietta, Elisabeth M / Hebbar, Nikhil /
    Velasquez, M Paulina / Mullighan, Charles G

    Cell reports

    2023  Volume 42, Issue 7, Page(s) 112804

    Abstract: The bone marrow microenvironment (BME) drives drug resistance in acute lymphoblastic leukemia (ALL) through leukemic cell interactions with bone marrow (BM) niches, but the underlying mechanisms remain unclear. Here, we show that the interaction between ... ...

    Abstract The bone marrow microenvironment (BME) drives drug resistance in acute lymphoblastic leukemia (ALL) through leukemic cell interactions with bone marrow (BM) niches, but the underlying mechanisms remain unclear. Here, we show that the interaction between ALL and mesenchymal stem cells (MSCs) through integrin β1 induces an epithelial-mesenchymal transition (EMT)-like program in MSC-adherent ALL cells, resulting in drug resistance and enhanced survival. Moreover, single-cell RNA sequencing analysis of ALL-MSC co-culture identifies a hybrid cluster of MSC-adherent ALL cells expressing both B-ALL and MSC signature genes, orchestrated by a WNT/β-catenin-mediated EMT-like program. Blockade of interaction between β-catenin and CREB binding protein impairs the survival and drug resistance of MSC-adherent ALL cells in vitro and results in a reduction in leukemic burden in vivo. Targeting of this WNT/β-catenin-mediated EMT-like program is a potential therapeutic approach to overcome cell extrinsically acquired drug resistance in ALL.
    MeSH term(s) Humans ; Epithelial-Mesenchymal Transition ; beta Catenin ; Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy ; Coculture Techniques ; Drug Resistance ; Cell Proliferation ; Tumor Microenvironment
    Chemical Substances beta Catenin
    Language English
    Publishing date 2023-07-14
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 2649101-1
    ISSN 2211-1247 ; 2211-1247
    ISSN (online) 2211-1247
    ISSN 2211-1247
    DOI 10.1016/j.celrep.2023.112804
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Genetic control of quiescence in hematopoietic stem cells.

    Yamada, Takeshi / Park, Chun Shik / Lacorazza, H Daniel

    Cell cycle (Georgetown, Tex.)

    2013  Volume 12, Issue 15, Page(s) 2376–2383

    Abstract: Cellular quiescence is a reversible cell cycle arrest that is poised to re-enter the cell cycle in response to a combination of cell-intrinsic factors and environmental cues. In hematopoietic stem cells, a coordinated balance between quiescence and ... ...

    Abstract Cellular quiescence is a reversible cell cycle arrest that is poised to re-enter the cell cycle in response to a combination of cell-intrinsic factors and environmental cues. In hematopoietic stem cells, a coordinated balance between quiescence and differentiating proliferation ensures longevity and prevents both genetic damage and stem cell exhaustion. However, little is known about how all these processes are integrated at the molecular level. We will briefly review the environmental and intrinsic control of stem cell quiescence and discuss a new model that involves a protein-to-protein interaction between G0S2 and the phospho-nucleoprotein nucleolin in the cytosol.
    MeSH term(s) Amino Acid Sequence ; Animals ; Cell Cycle Checkpoints/genetics ; Cell Cycle Proteins/chemistry ; Cell Cycle Proteins/physiology ; Cell Proliferation ; Gene Expression Regulation ; Hematopoietic Stem Cells/physiology ; Humans ; Molecular Sequence Data ; Phosphoproteins/metabolism ; Protein Binding ; Protein Interaction Domains and Motifs ; RNA-Binding Proteins/metabolism ; Nucleolin
    Chemical Substances Cell Cycle Proteins ; G0S2 protein, human ; Phosphoproteins ; RNA-Binding Proteins
    Language English
    Publishing date 2013-06-28
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Review
    ZDB-ID 2146183-1
    ISSN 1551-4005 ; 1538-4101 ; 1554-8627
    ISSN (online) 1551-4005
    ISSN 1538-4101 ; 1554-8627
    DOI 10.4161/cc.25416
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: An ELF4 hypomorphic variant results in NK cell deficiency.

    Salinas, Sandra Andrea / Mace, Emily M / Conte, Matilde I / Park, Chun Shik / Li, Yu / Rosario-Sepulveda, Joshua I / Mahapatra, Sanjana / Moore, Emily K / Hernandez, Evelyn R / Chinn, Ivan K / Reed, Abigail E / Lee, Barclay J / Frumovitz, Alexander / Gibbs, Richard A / Posey, Jennifer E / Forbes Satter, Lisa R / Thatayatikom, Akaluck / Allenspach, Eric J / Wensel, Theodore G /
    Lupski, James R / Lacorazza, H Daniel / Orange, Jordan S

    JCI insight

    2022  Volume 7, Issue 23

    Abstract: NK cell deficiencies (NKD) are a type of primary immune deficiency in which the major immunologic abnormality affects NK cell number, maturity, or function. Since NK cells contribute to immune defense against virally infected cells, patients with NKD ... ...

    Abstract NK cell deficiencies (NKD) are a type of primary immune deficiency in which the major immunologic abnormality affects NK cell number, maturity, or function. Since NK cells contribute to immune defense against virally infected cells, patients with NKD experience higher susceptibility to chronic, recurrent, and fatal viral infections. An individual with recurrent viral infections and mild hypogammaglobulinemia was identified to have an X-linked damaging variant in the transcription factor gene ELF4. The variant does not decrease expression but disrupts ELF4 protein interactions and DNA binding, reducing transcriptional activation of target genes and selectively impairing ELF4 function. Corroborating previous murine models of ELF4 deficiency (Elf4-/-) and using a knockdown human NK cell line, we determined that ELF4 is necessary for normal NK cell development, terminal maturation, and function. Through characterization of the NK cells of the proband, expression of the proband's variant in Elf4-/- mouse hematopoietic precursor cells, and a human in vitro NK cell maturation model, we established this ELF4 variant as a potentially novel cause of NKD.
    MeSH term(s) Animals ; Humans ; Mice ; DNA-Binding Proteins/genetics ; Killer Cells, Natural ; Transcription Factors/genetics
    Chemical Substances DNA-Binding Proteins ; ELF4 protein, human ; Transcription Factors ; Elf4 protein, mouse
    Language English
    Publishing date 2022-12-08
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Research Support, N.I.H., Extramural
    ISSN 2379-3708
    ISSN (online) 2379-3708
    DOI 10.1172/jci.insight.155481
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: G0S2 modulates homeostatic proliferation of naïve CD8⁺ T cells and inhibits oxidative phosphorylation in mitochondria.

    Lee, Ping-Hsien / Yamada, Takeshi / Park, Chun Shik / Shen, Ye / Puppi, Monica / Lacorazza, H Daniel

    Immunology and cell biology

    2015  Volume 93, Issue 7, Page(s) 605–615

    Abstract: Since its discovery, diverse functions have been attributed to the G0/G1 switch gene 2 (G0S2), from lipid metabolism to control of cell proliferation. Our group showed for the first time that G0S2 promotes quiescence in hematopoietic stem cells by ... ...

    Abstract Since its discovery, diverse functions have been attributed to the G0/G1 switch gene 2 (G0S2), from lipid metabolism to control of cell proliferation. Our group showed for the first time that G0S2 promotes quiescence in hematopoietic stem cells by interacting with and retaining nucleolin around the nucleus. Herein, we report the role of G0S2 in the differentiation and function of CD8(+) T cells examined in mice with an embryonic deletion of the G0s2 gene. G0S2 expression in naïve CD8(+) T cells decreased immediately after T-cell receptor activation downstream of the mitogen-activated protein kinase, calcium/calmodulin, phosphatidylinositol 3'-kinase and mammalian target of rapamycin pathways. Surprisingly, G0S2-null naïve CD8(+) T cells displayed increased basal and spare respiratory capacity that was not associated with increased mitochondrial biogenesis but with increased phosphorylation of AMP-activated protein kinase α. Naïve CD8(+) T cells showed increased proliferation in response to in vitro activation and in vivo lymphopenia; however, naïve CD8(+) T cells expressing the OT-1 transgene exhibited normal differentiation of naïve cells to effector and memory CD8(+) T cells upon infection with Listeria monocytogenes in a wild-type or a G0s2-null environment, with increased circulating levels of free fatty acids. Collectively, our results suggest that G0S2 inhibits energy production by oxidative phosphorylation to fine-tune proliferation in homeostatic conditions.
    MeSH term(s) Animals ; CD8-Positive T-Lymphocytes/cytology ; CD8-Positive T-Lymphocytes/immunology ; CD8-Positive T-Lymphocytes/metabolism ; Cell Cycle Proteins/deficiency ; Cell Cycle Proteins/genetics ; Cell Cycle Proteins/physiology ; Cell Division ; Cells, Cultured ; Cyclic AMP-Dependent Protein Kinases/metabolism ; Fatty Acids, Nonesterified/blood ; Female ; Genes, Reporter ; Homeostasis ; Listeria monocytogenes ; Listeriosis/immunology ; Listeriosis/metabolism ; Lymphocyte Activation ; Lymphopenia/immunology ; Lymphopenia/metabolism ; Lymphopoiesis ; Male ; Mice ; Mice, Inbred C57BL ; Mice, Transgenic ; Mitochondria/metabolism ; Oxidative Phosphorylation ; Phosphorylation ; Protein Processing, Post-Translational ; Receptors, Antigen, T-Cell/immunology ; Signal Transduction ; Specific Pathogen-Free Organisms
    Chemical Substances Cell Cycle Proteins ; Fatty Acids, Nonesterified ; G0S2 protein, mouse ; Receptors, Antigen, T-Cell ; Cyclic AMP-Dependent Protein Kinases (EC 2.7.11.11)
    Language English
    Publishing date 2015-02-10
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 284057-1
    ISSN 1440-1711 ; 0818-9641
    ISSN (online) 1440-1711
    ISSN 0818-9641
    DOI 10.1038/icb.2015.9
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: G0S2 inhibits the proliferation of K562 cells by interacting with nucleolin in the cytosol.

    Yamada, Takeshi / Park, Chun Shik / Shen, Ye / Rabin, Karen R / Lacorazza, H Daniel

    Leukemia research

    2013  Volume 38, Issue 2, Page(s) 210–217

    Abstract: G0/G1 switch gene 2 (G0S2) is a basic protein with ill-defined function that inhibits the proliferation of hematopoietic stem cells. Herein, we show that treatment of K562 cells with 5-azacytidine (5-Aza) resulted in a 24-fold increase in G0S2 expression ...

    Abstract G0/G1 switch gene 2 (G0S2) is a basic protein with ill-defined function that inhibits the proliferation of hematopoietic stem cells. Herein, we show that treatment of K562 cells with 5-azacytidine (5-Aza) resulted in a 24-fold increase in G0S2 expression and a reduction in cell growth. Conversely, gene demethylation in the presence of G0S2-specific shRNA restored proliferation, further supporting an inhibitory role for G0S2 in cell proliferation. Elevated levels of G0S2 inhibited the division of K562 cells by sequestering the nucleolar phosphoprotein nucleolin in the cytosol. G0S2 inhibited the proliferation of leukemia cells in vivo in xenograft models. Collectively, our data identify a new mechanism that controls proliferation in K562 cells, suggesting a possible tumor suppressor function in leukemia cells.
    MeSH term(s) Animals ; Cell Cycle Proteins/antagonists & inhibitors ; Cell Cycle Proteins/physiology ; Cell Proliferation/drug effects ; Cells, Cultured ; Cytosol/metabolism ; Genes, Tumor Suppressor/physiology ; HL-60 Cells ; Humans ; K562 Cells ; Mice ; Mice, Nude ; Phosphoproteins/metabolism ; Protein Binding/drug effects ; RNA, Small Interfering/pharmacology ; RNA-Binding Proteins/metabolism ; Nucleolin
    Chemical Substances Cell Cycle Proteins ; G0S2 protein, human ; Phosphoproteins ; RNA, Small Interfering ; RNA-Binding Proteins
    Language English
    Publishing date 2013-10-14
    Publishing country England
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 752396-8
    ISSN 1873-5835 ; 0145-2126
    ISSN (online) 1873-5835
    ISSN 0145-2126
    DOI 10.1016/j.leukres.2013.10.006
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: Differential roles of KLF4 in the development and differentiation of CD8+ T cells.

    Mamonkin, Maksim / Shen, Ye / Lee, Ping-Hsien / Puppi, Monica / Park, Chun Shik / Lacorazza, H Daniel

    Immunology letters

    2013  Volume 156, Issue 1-2, Page(s) 94–101

    Abstract: The transcription factor Krüppel-like factor 4 (KLF4) can activate or repress gene expression in a cell-context dependent manner. We have previously shown that KLF4 inhibits the proliferation of naïve CD8(+) T cells in vitro downstream of the ... ...

    Abstract The transcription factor Krüppel-like factor 4 (KLF4) can activate or repress gene expression in a cell-context dependent manner. We have previously shown that KLF4 inhibits the proliferation of naïve CD8(+) T cells in vitro downstream of the transcription factor ELF4. In this work, we describe a novel role of KLF4 in the differentiation of CD8(+) T cells upon infection. Loss of KLF4 had minimal effect on thymic T cell development and distribution of mature T cells in the spleen, blood, and lymph nodes. KLF4-deficient naïve CD8(+) T cells also displayed normal homeostatic proliferation upon adoptive transfer into lymphopenic hosts. However, activation of KLF4-deficient naïve CD8(+) T cells by in vitro TCR crosslink and co-stimulation resulted in increased proliferation. Furthermore, naïve KLF4-deficient OT-I CD8(+) T cells generated increased numbers of functional memory CD8(+) T cells compared to wild type OT-I CD8(+) T cells co-injected in the same recipient in both primary and recall responses to Listeria monocytogenes-OVA. Collectively, our data demonstrate that KLF4 regulates differentiation of functional memory CD8(+) T cells while sparing development and homeostasis of naïve CD8(+) T cells.
    MeSH term(s) Adoptive Transfer ; Animals ; CD8-Positive T-Lymphocytes/immunology ; CD8-Positive T-Lymphocytes/metabolism ; Cell Differentiation/genetics ; Cell Differentiation/immunology ; Cell Proliferation ; Flow Cytometry ; Homeostasis/immunology ; Immunologic Memory/immunology ; Kruppel-Like Transcription Factors/genetics ; Kruppel-Like Transcription Factors/immunology ; Kruppel-Like Transcription Factors/metabolism ; Listeria monocytogenes/immunology ; Listeriosis/genetics ; Listeriosis/immunology ; Listeriosis/microbiology ; Mice ; Mice, Inbred C57BL ; Mice, Inbred Strains ; Mice, Knockout ; Mice, Transgenic ; Receptors, Antigen, T-Cell/immunology
    Chemical Substances GKLF protein ; Kruppel-Like Transcription Factors ; Receptors, Antigen, T-Cell
    Language English
    Publishing date 2013-09-25
    Publishing country Netherlands
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ZDB-ID 445150-8
    ISSN 1879-0542 ; 0165-2478
    ISSN (online) 1879-0542
    ISSN 0165-2478
    DOI 10.1016/j.imlet.2013.09.008
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: The cytosolic protein G0S2 maintains quiescence in hematopoietic stem cells.

    Yamada, Takeshi / Park, Chun Shik / Burns, Audrea / Nakada, Daisuke / Lacorazza, H Daniel

    PloS one

    2012  Volume 7, Issue 5, Page(s) e38280

    Abstract: Bone marrow hematopoietic stem cells (HSCs) balance proliferation and differentiation by integrating complex transcriptional and post-translational mechanisms regulated by cell intrinsic and extrinsic factors. We found that transcripts of G(0)/G(1) ... ...

    Abstract Bone marrow hematopoietic stem cells (HSCs) balance proliferation and differentiation by integrating complex transcriptional and post-translational mechanisms regulated by cell intrinsic and extrinsic factors. We found that transcripts of G(0)/G(1) switch gene 2 (G0S2) are enriched in lineage(-) Sca-1(+) c-kit(+) (LSK) CD150(+) CD48(-) CD41(-) cells, a population highly enriched for quiescent HSCs, whereas G0S2 expression is suppressed in dividing LSK CD150(+) CD48(-) cells. Gain-of-function analyses using retroviral expression vectors in bone marrow cells showed that G0S2 localizes to the mitochondria, endoplasmic reticulum, and early endosomes in hematopoietic cells. Co-transplantation of bone marrow cells transduced with the control or G0S2 retrovirus led to increased chimerism of G0S2-overexpressing cells in femurs, although their contribution to the blood was reduced. This finding was correlated with increased quiescence in G0S2-overexpressing HSCs (LSK CD150(+) CD48(-)) and progenitor cells (LS(-)K). Conversely, silencing of endogenous G0S2 expression in bone marrow cells increased blood chimerism upon transplantation and promoted HSC cell division, supporting an inhibitory role for G0S2 in HSC proliferation. A proteomic study revealed that the hydrophobic domain of G0S2 interacts with a domain of nucleolin that is rich in arginine-glycine-glycine repeats, which results in the retention of nucleolin in the cytosol. We showed that this cytosolic retention of nucleolin occurs in resting, but not proliferating, wild-type LSK CD150(+) CD48(-) cells. Collectively, we propose a novel model of HSC quiescence in which elevated G0S2 expression can sequester nucleolin in the cytosol, precluding its pro-proliferation functions in the nucleolus.
    MeSH term(s) Animals ; Blood Cells/metabolism ; Cell Cycle ; Cell Cycle Proteins/chemistry ; Cell Cycle Proteins/genetics ; Cell Cycle Proteins/metabolism ; Cytosol/metabolism ; Gene Expression Regulation ; HEK293 Cells ; Hematopoietic Stem Cells/cytology ; Hematopoietic Stem Cells/metabolism ; Humans ; Hydrophobic and Hydrophilic Interactions ; Mice ; NIH 3T3 Cells ; Phosphoproteins/metabolism ; Protein Structure, Tertiary ; RNA-Binding Proteins/metabolism ; Ribonucleoproteins/metabolism ; Nucleolin
    Chemical Substances Cell Cycle Proteins ; G0S2 protein, mouse ; Phosphoproteins ; RNA-Binding Proteins ; Ribonucleoproteins
    Language English
    Publishing date 2012-05-31
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 2267670-3
    ISSN 1932-6203 ; 1932-6203
    ISSN (online) 1932-6203
    ISSN 1932-6203
    DOI 10.1371/journal.pone.0038280
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top