1
|
Ead JK, Armstrong DG. Granulocyte-macrophage colony-stimulating factor: Conductor of the wound healing orchestra? Int Wound J 2023; 20:1229-1234. [PMID: 36632762 PMCID: PMC10031218 DOI: 10.1111/iwj.13919] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/25/2022] [Indexed: 01/13/2023] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a glycoprotein and is derived from both hemopoietic and nonhemopoietic sources which exert immunomodulatory properties. Various theories have been proposed to explain why some wounds become chronic and non-healing. Generalized suppression of inflammation locally or systemically may impede the body's physiological healing response by crippling the activity of reparative cells within the wound ecosystem. Thus, highlighting the importance of promoting host-directed therapeutics with immunomodulatory properties. The temporal and spatial expression of GM-CSF and GM-CSF receptors in the integumentary system suggests that epithelial-derived GM-CSF functions in an autocrine/paracrine manner. This may positively affect wound healing physiology via local inflammatory regulation promoting macrophage survival. Although diabetes negatively affects multiple aspects of wound healing GM-CSF activation is particularly impacted. Compared to acute/healthy wounds diabetic foot ulcers (DFU) only partially activate GM-CSF activity. There is a deleterious chain of events associated with this unfortunate sequala. DFUs also have a high proportion of monocytes and an absence of activated macrophages which results in an impaired inflammatory response. This may potentially serve as a vital point for GM-CSF to act as a companion diagnostic/theragnostic modality to help modulate the inflammatory response in wound healing. Correcting macrophage immune dysfunction with exogenous GM-CSF may help restore the immune balance in the wound ecosystem and jumpstart the wound healing cascade. Thus, the recognized beneficial role of GM-CSF in immune regulation across many studies provides a rationale for the initiation of the ongoing randomized controlled trials using GM-CSF.
Collapse
Affiliation(s)
- J. Karim Ead
- Department of SurgeryUSC Keck School of MedicineLos AngelesUSA
| | | |
Collapse
|
2
|
Tamai M, Fujisawa S, Nguyen TTT, Komatsu C, Kagami K, Kamimoto K, Omachi K, Kasai S, Harama D, Watanabe A, Akahane K, Goi K, Naka K, Kaname T, Teshima T, Inukai T. Creation of Philadelphia chromosome by CRISPR/Cas9-mediated double cleavages on BCR and ABL1 genes as a model for initial event in leukemogenesis. Cancer Gene Ther 2023; 30:38-50. [PMID: 35999358 PMCID: PMC9842507 DOI: 10.1038/s41417-022-00522-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/27/2022] [Accepted: 08/04/2022] [Indexed: 01/21/2023]
Abstract
The Philadelphia (Ph) chromosome was the first translocation identified in leukemia. It is supposed to be generated by aberrant ligation between two DNA double-strand breaks (DSBs) at the BCR gene located on chromosome 9q34 and the ABL1 gene located on chromosome 22q11. Thus, mimicking the initiation process of translocation, we induced CRISPR/Cas9-mediated DSBs simultaneously at the breakpoints of the BCR and ABL1 genes in a granulocyte-macrophage colony-stimulating factor (GM-CSF) dependent human leukemia cell line. After transfection of two single guide RNAs (sgRNAs) targeting intron 13 of the BCR gene and intron 1 of the ABL1 gene, a factor-independent subline was obtained. In the subline, p210 BCR::ABL1 and its reciprocal ABL1::BCR fusions were generated as a result of balanced translocation corresponding to the Ph chromosome. Another set of sgRNAs targeting intron 1 of the BCR gene and intron 1 of the ABL1 gene induced a factor-independent subline expressing p190 BCR::ABL1. Both p210 and p190 BCR::ABL1 induced factor-independent growth by constitutively activating intracellular signaling pathways for transcriptional regulation of cell cycle progression and cell survival that are usually regulated by GM-CSF. These observations suggested that simultaneous DSBs at the BCR and ABL1 gene breakpoints are initiation events for oncogenesis in Ph+ leukemia. (200/200 words).
Collapse
Affiliation(s)
- Minori Tamai
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan.
| | - Shinichi Fujisawa
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Hokkaido, Japan
| | - Thao T T Nguyen
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Chiaki Komatsu
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Keiko Kagami
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kenji Kamimoto
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St Louis, MO, USA
| | - Kohei Omachi
- Division of Nephrology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Shin Kasai
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Daisuke Harama
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Atsushi Watanabe
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Koshi Akahane
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kumiko Goi
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kazuhito Naka
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Tadashi Kaname
- Department of Genome Medicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Takanori Teshima
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Hokkaido, Japan
| | - Takeshi Inukai
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
3
|
Grant AH, Estrada A, Ayala-Marin YM, Alvidrez-Camacho AY, Rodriguez G, Robles-Escajeda E, Cadena-Medina DA, Rodriguez AC, Kirken RA. The Many Faces of JAKs and STATs Within the COVID-19 Storm. Front Immunol 2021; 12:690477. [PMID: 34326843 PMCID: PMC8313986 DOI: 10.3389/fimmu.2021.690477] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
The positive-sense single stranded RNA virus, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), resulted in a global pandemic with horrendous health and economic consequences not seen in a century. At a finer scale, immunologically, many of these devastating effects by SARS-CoV-2 can be traced to a "cytokine storm" resulting in the simultaneous activation of Janus Kinases (JAKs) and Signal Transducers and Activators of Transcription (STAT) proteins downstream of the many cytokine receptor families triggered by elevated cytokines found in Coronavirus Disease 2019 (COVID-19). In this report, cytokines found in the storm are discussed in relation to the JAK-STAT pathway in response to SARS-CoV-2 and the lessons learned from RNA viruses and previous Coronaviruses (CoVs). Therapeutic strategies to counteract the SARS-CoV-2 mediated storm are discussed with an emphasis on cell signaling and JAK inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Robert A. Kirken
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, United States
| |
Collapse
|
4
|
Bisht K, Brunck ME, Matsumoto T, McGirr C, Nowlan B, Fleming W, Keech T, Magor G, Perkins AC, Davies J, Walkinshaw G, Flippin L, Winkler IG, Levesque JP. HIF prolyl hydroxylase inhibitor FG-4497 enhances mouse hematopoietic stem cell mobilization via VEGFR2/KDR. Blood Adv 2019; 3:406-418. [PMID: 30733301 PMCID: PMC6373754 DOI: 10.1182/bloodadvances.2018017566] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 01/06/2019] [Indexed: 02/06/2023] Open
Abstract
In normoxia, hypoxia-inducible transcription factors (HIFs) are rapidly degraded within the cytoplasm as a consequence of their prolyl hydroxylation by oxygen-dependent prolyl hydroxylase domain (PHD) enzymes. We have previously shown that hematopoietic stem and progenitor cells (HSPCs) require HIF-1 for effective mobilization in response to granulocyte colony-stimulating factor (G-CSF) and CXCR4 antagonist AMD3100/plerixafor. Conversely, HIF PHD inhibitors that stabilize HIF-1 protein in vivo enhance HSPC mobilization in response to G-CSF or AMD3100 in a cell-intrinsic manner. We now show that extrinsic mechanisms involving vascular endothelial growth factor receptor-2 (VEGFR2), via bone marrow (BM) endothelial cells, are also at play. PTK787/vatalanib, a tyrosine kinase inhibitor selective for VEGFR1 and VEGFR2, and neutralizing anti-VEGFR2 monoclonal antibody DC101 blocked enhancement of HSPC mobilization by FG-4497. VEGFR2 was absent on mesenchymal and hematopoietic cells and was detected only in Sca1+ endothelial cells in the BM. We propose that HIF PHD inhibitor FG-4497 enhances HSPC mobilization by stabilizing HIF-1α in HSPCs as previously demonstrated, as well as by activating VEGFR2 signaling in BM endothelial cells, which facilitates HSPC egress from the BM into the circulation.
Collapse
Affiliation(s)
- Kavita Bisht
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Marion E Brunck
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Taichi Matsumoto
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
- Faculty of Pharmacological Sciences, Fukuoka University, Fukuoka, Japan
| | - Crystal McGirr
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Bianca Nowlan
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Whitney Fleming
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Thomas Keech
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Graham Magor
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Andrew C Perkins
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Julie Davies
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | | | | | - Ingrid G Winkler
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| | - Jean-Pierre Levesque
- Cancer Care and Biology Program, Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| |
Collapse
|
5
|
Laulicht F, Brocato J, Cartularo L, Vaughan J, Wu F, Kluz T, Sun H, Oksuz BA, Shen S, Peana M, Medici S, Zoroddu MA, Costa M. Tungsten-induced carcinogenesis in human bronchial epithelial cells. Toxicol Appl Pharmacol 2015; 288:33-9. [PMID: 26164860 PMCID: PMC4579035 DOI: 10.1016/j.taap.2015.07.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/30/2015] [Accepted: 07/02/2015] [Indexed: 12/30/2022]
Abstract
Metals such as arsenic, cadmium, beryllium, and nickel are known human carcinogens; however, other transition metals, such as tungsten (W), remain relatively uninvestigated with regard to their potential carcinogenic activity. Tungsten production for industrial and military applications has almost doubled over the past decade and continues to increase. Here, for the first time, we demonstrate tungsten's ability to induce carcinogenic related endpoints including cell transformation, increased migration, xenograft growth in nude mice, and the activation of multiple cancer-related pathways in transformed clones as determined by RNA sequencing. Human bronchial epithelial cell line (Beas-2B) exposed to tungsten developed carcinogenic properties. In a soft agar assay, tungsten-treated cells formed more colonies than controls and the tungsten-transformed clones formed tumors in nude mice. RNA-sequencing data revealed that the tungsten-transformed clones altered the expression of many cancer-associated genes when compared to control clones. Genes involved in lung cancer, leukemia, and general cancer genes were deregulated by tungsten. Taken together, our data show the carcinogenic potential of tungsten. Further tests are needed, including in vivo and human studies, in order to validate tungsten as a carcinogen to humans.
Collapse
Affiliation(s)
- Freda Laulicht
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, NY 10987, USA
| | - Jason Brocato
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, NY 10987, USA
| | - Laura Cartularo
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, NY 10987, USA
| | - Joshua Vaughan
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, NY 10987, USA
| | - Feng Wu
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, NY 10987, USA
| | - Thomas Kluz
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, NY 10987, USA
| | - Hong Sun
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, NY 10987, USA
| | - Betul Akgol Oksuz
- Genome Technology Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - Steven Shen
- Center for Health Informatics and Bioinformatics, New York University Langone Medical Center, New York, NY 10016, USA
| | - Massimiliano Peana
- Department of Chemistry and Pharmacy, University of Sassari, Sassari, Italy
| | - Serenella Medici
- Department of Chemistry and Pharmacy, University of Sassari, Sassari, Italy
| | | | - Max Costa
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, NY 10987, USA.
| |
Collapse
|
6
|
Firas J, Liu X, Nefzger CM, Polo JM. GM-CSF and MEF-conditioned media support feeder-free reprogramming of mouse granulocytes to iPS cells. Differentiation 2014; 87:193-9. [PMID: 25015842 DOI: 10.1016/j.diff.2014.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 05/24/2014] [Accepted: 05/29/2014] [Indexed: 01/30/2023]
Abstract
Induced pluripotent stem cells (iPSCs) are characterised by their ability to differentiate into any cell type of the body. Accordingly, iPSCs possess immense potential for disease modelling, pharmaceutical screening and autologous cell therapies. The most common source of iPSCs derivation is skin fibroblasts. However, from a clinical point of view, skin fibroblasts may not be ideal, as invasive procedures such as skin biopsies are required for their extraction. Moreover, fibroblasts are highly heterogeneous with a poorly defined developmental pathway, which makes studying reprogramming mechanistics difficult. Granulocytes, on the other hand, are easily obtainable, their developmental pathway has been extensively studied and fluorescence activated cell sorting allows for the isolation of these cells at high purity; thus iPSCs derivation from granulocytes could provide an alternative to fibroblast-derived iPSCs. Previous studies succeeded in producing iPSC colonies from mouse granulocytes but with the use of a mitotically inactivated feeder layer, restricting their use for studying reprogramming mechanistics. As granulocytes display poor survival under culture conditions, we investigated the influence of haematopoietic cytokines to stabilise this cell type in vitro and allow for reprogramming in the absence of a feeder layer. Our results show that treatment with MEF-conditioned media and/or initial exposure to GM-CSF allows for reprogramming of granulocytes under feeder-free conditions. This work can serve as a basis for future work aimed at dissecting the reprogramming mechanism as well as obtaining large numbers of iPSCs from a clinically relevant cell source.
Collapse
Affiliation(s)
- Jaber Firas
- Department of Anatomy and Developmental Biology, Monash University, Clayton 3800, Victoria, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Xiaodong Liu
- Department of Anatomy and Developmental Biology, Monash University, Clayton 3800, Victoria, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Christian M Nefzger
- Department of Anatomy and Developmental Biology, Monash University, Clayton 3800, Victoria, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton 3800, Victoria, Australia.
| | - Jose M Polo
- Department of Anatomy and Developmental Biology, Monash University, Clayton 3800, Victoria, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton 3800, Victoria, Australia.
| |
Collapse
|
7
|
Yamamoto T, Mase K, Sawada H. Diapause prevention effect of Bombyx mori by dimethyl sulfoxide. PLoS One 2013; 8:e64124. [PMID: 23675522 PMCID: PMC3652847 DOI: 10.1371/journal.pone.0064124] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 04/10/2013] [Indexed: 01/04/2023] Open
Abstract
HCl treatment has been, for about 80 years, the primary method for the prevention of entry into embryonic diapauses of Bombyx mori. This is because no method is as effective as the HCl treatment. In this study, we discovered that dimethyl sulfoxide (DMSO) prevented entry into the diapause of the silkworm, Bombyx mori. The effect of diapause prevention was 78% as a result of treatment with 100% DMSO concentration, and the effect was comparable to that of the HCl treatment. In contrast, in the case of non-diapause eggs, hatchability was decreased by DMSO in a concentration-dependent manner. The effect of DMSO was restricted within 24 hours after oviposition of diapause eggs, and the critical period was slightly shorter than the effective period of the HCl treatment. DMSO analogs, such as dimethyl formamide (DMF) and dimethyl sulfide (DMS), did little preventive effect against the diapause. Furthermore, we also investigated the permeation effects of chemical compounds by DMSO. When treated with an inhibitor of protein kinase CK2 (CK2) dissolved in DMSO, the prevention rate of the diapause was less than 40%. This means that the inhibition effect by the CK2 inhibitor was the inhibition of embryonic development after diapause prevention by DMSO. These data suggest that DMSO has the effects of preventing from entering into the diapause and permeation of chemicals into diapause eggs.
Collapse
Affiliation(s)
- Takayuki Yamamoto
- Division of Biology, College of Liberal Arts and Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Keisuke Mase
- College of Humanities and Sciences, Nihon University, Setagaya-ku, Tokyo, Japan
| | - Hiroshi Sawada
- College of Humanities and Sciences, Nihon University, Setagaya-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
8
|
Kanayasu-Toyoda T, Ishii-Watabe A, Suzuki T, Oshizawa T, Yamaguchi T. A New Role of Thrombopoietin Enhancing ex Vivo Expansion of Endothelial Precursor Cells Derived from AC133-positive Cells. J Biol Chem 2007; 282:33507-33514. [PMID: 17827152 DOI: 10.1074/jbc.m703919200] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We previously reported that CD31(bright) cells, which were sorted from cultured AC133(+) cells of adult peripheral blood cells, differentiated more efficiently into endothelial cells than CD31(+) cells or CD31(-) cells, suggesting that CD31(bright) cells may be endothelial precursor cells. In this study, we found that CD31(bright) cells have a strong ability to release cytokines. The mixture of vascular endothelial growth factor (VEGF), thrombopoietin (TPO), and stem cell factor stimulated ex vivo expansion of the total cell number from cultured AC133(+) cells of adult peripheral blood cells and cord blood cells, resulting in incrementation of the adhesion cells, in which endothelial nitric oxide synthase and kinase insert domain-containing receptor were positive. Moreover, the mixture of VEGF and TPO increased the CD31(bright) cell population when compared with VEGF alone or the mixture of VEGF and stem cell factor. These data suggest that TPO is an important growth factor that can promote endothelial precursor cells expansion ex vivo.
Collapse
Affiliation(s)
- Toshie Kanayasu-Toyoda
- Department of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kamiyoga 1-18-1, Setagayaku, Tokyo, 158-8501, Japan
| | - Akiko Ishii-Watabe
- Department of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kamiyoga 1-18-1, Setagayaku, Tokyo, 158-8501, Japan
| | - Takayoshi Suzuki
- Department of Cellular and Gene Therapy Products, National Institute of Health Sciences, Kamiyoga 1-18-1, Setagayaku, Tokyo 158-8501, Japan
| | - Tadashi Oshizawa
- Department of Cellular and Gene Therapy Products, National Institute of Health Sciences, Kamiyoga 1-18-1, Setagayaku, Tokyo 158-8501, Japan
| | - Teruhide Yamaguchi
- Department of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kamiyoga 1-18-1, Setagayaku, Tokyo, 158-8501, Japan.
| |
Collapse
|
9
|
Kanayasu-Toyoda T, Suzuki T, Oshizawa T, Uchida E, Hayakawa T, Yamaguchi T. Granulocyte colony-stimulating factor promotes the translocation of protein kinase Ciota in neutrophilic differentiation cells. J Cell Physiol 2007; 211:189-96. [PMID: 17133348 DOI: 10.1002/jcp.20930] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Previously, we suggested that the phosphatidylinositol 3-kinase (PI3K)-p70 S6 kinase (p70 S6K) pathway plays an important role in granulocyte colony-stimulating factor (G-CSF)-dependent enhancement of the neutrophilic differentiation and proliferation of HL-60 cells. While atypical protein kinase C (PKC) has been reported to be a regulator of p70 S6K, abundant expression of PKCiota was observed in myeloid and lymphoid cells. Therefore, we analyzed the participation of PKCiota in G-CSF-dependent proliferation. The maximum stimulation of PKCiota was observed from 15 to 30 min after the addition of G-CSF. From 5 to 15 min into this lag time, PKCiota was found to translocate from the nucleus to the membrane. At 30 min it re-translocated to the cytosol. This dynamic translocation of PKCiota was also observed in G-CSF-stimulated myeloperoxidase-positive cells differentiated from cord blood cells. Small interfering RNA for PKCiota inhibited G-CSF-induced proliferation and the promotion of neutrophilic differentiation of HL-60 cells. These data indicate that the G-CSF-induced dynamic translocation and activation processes of PKCiota are important to neutrophilic proliferation.
Collapse
Affiliation(s)
- Toshie Kanayasu-Toyoda
- Division of Cellular and Gene Therapy Products, National Institute of Health Sciences, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
10
|
Yost CC, Denis MM, Lindemann S, Rubner FJ, Marathe GK, Buerke M, McIntyre TM, Weyrich AS, Zimmerman GA. Activated polymorphonuclear leukocytes rapidly synthesize retinoic acid receptor-alpha: a mechanism for translational control of transcriptional events. J Exp Med 2004; 200:671-80. [PMID: 15337793 PMCID: PMC2212748 DOI: 10.1084/jem.20040224] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2004] [Accepted: 07/27/2004] [Indexed: 11/04/2022] Open
Abstract
In addition to releasing preformed granular proteins, polymorphonuclear leukocytes (PMNs) synthesize chemokines and other factors under transcriptional control. Here we demonstrate that PMNs express an inducible transcriptional modulator by signal-dependent activation of specialized mechanisms that regulate messenger RNA (mRNA) translation. HL-60 myelocytic cells differentiated to surrogate PMNs respond to activation by platelet activating factor by initiating translation and with appearance of specific mRNA transcripts in polyribosomes. cDNA array analysis of the polyribosome fraction demonstrated that retinoic acid receptor (RAR)-alpha, a transcription factor that controls the expression of multiple genes, is one of the polyribosome-associated transcripts. Quiescent surrogate HL60 PMNs and primary human PMNs contain constitutive message for RAR-alpha but little or no protein. RAR-alpha protein is rapidly synthesized in response to platelet activating factor under the control of a specialized translational regulator, mammalian target of rapamycin, and is blocked by the therapeutic macrolide rapamycin, events consistent with features of the 5' untranslated region of the transcript. Newly synthesized RAR-alpha modulates production of interleukin-8. Rapid expression of a transcription factor under translational control is a previously unrecognized mechanism in human PMNs that indicates unexpected diversity in gene regulation in this critical innate immune effector cell.
Collapse
Affiliation(s)
- Christian C Yost
- Department of Pediatrics, Eccles Institute of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Gomez-Cambronero J, Frye T, Baumann M. Ribosomal p70S6K basal activity increases upon induction of differentiation of myelomonocytic leukemic cell lines HL60, AML14 and MPD. Leuk Res 2004; 28:755-62. [PMID: 15158097 PMCID: PMC3082395 DOI: 10.1016/j.leukres.2003.11.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2003] [Accepted: 11/21/2003] [Indexed: 11/26/2022]
Abstract
The role of ribosomal p70S6K in the cell cycle has been studied extensively, and it is known that this enzyme is crucial for cell advancement through G(1). Conversely, the participation of p70S6K in cell differentiation is not well understood. We have studied the response of p70S6K to the cytokine granulocyte-macrophage colony stimulating factor (GM-CSF) in three differentiation-capable leukemic cell lines (MPD, AML-14 and HL-60) and in normal mature neutrophils. Immature leukemic cells starved for 16 h showed a robust ( approximately 3.5-fold over controls) p70S6K phosphorylation on T(421)/S(424) residues in response to an acute (5 min) 10 nM GM-CSF stimulation. On the other hand, cells that had been induced to differentiate and express granulocytic phenotypes, showed an increased ( approximately 6-fold) basal level of p70S6K T(421)/S(424) phosphorylation over immature cells, as well as an increased baseline tyrosyl phosphorylation of the GM-CSF receptor beta subunit (GM-CSF.Rbeta). However, the differentiated cells displayed a weak ( approximately 1.4-fold over controls) response to GM-CSF even at prolonged incubation times (20 min). In vitro p70S6K enzymatic activity paralleled p70S6K T(421)/S(424) phosphorylation in both high basal, unstimulated, levels in immature cells and a low degree of response to GM-CSF. Lastly, peripheral blood mature neutrophils had low basal GM-CSF.Rbeta and p70S6K activity, with both parameters being robustly stimulated following addition of GM-CSF, a situation in contrast with the cell lines, indicative perhaps of their incomplete terminal differentiation. In summary, these findings show the increase in basal phosphorylation of p70S6K upon granulocytic differentiation of myeloid leukemic cells and their responses to GM-CSF that are closely paralleled with tyrosyl phosphorylation of its receptor, and help in pointing to specific cell signaling molecules that are different in leukemic blasts from normal leukocytes.
Collapse
Affiliation(s)
- Julian Gomez-Cambronero
- Department of Physiology and Biophysics, School of Medicine, Wright State University, 3640 Colonel Glenn Highway, Dayton, OH 45435, USA.
| | | | | |
Collapse
|
12
|
Lehman JA, Calvo V, Gomez-Cambronero J. Mechanism of ribosomal p70S6 kinase activation by granulocyte macrophage colony-stimulating factor in neutrophils: cooperation of a MEK-related, THR421/SER424 kinase and a rapamycin-sensitive, m-TOR-related THR389 kinase. J Biol Chem 2003; 278:28130-8. [PMID: 12740386 DOI: 10.1074/jbc.m300376200] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We report here for the first time the detection of the ribosomal p70S6 kinase (p70S6K) in a hematopoietic cell, the neutrophil, and the stimulation of its enzymatic activity by granulocyte macrophage colony-stimulating factor (GM-CSF). GM-CSF modified the Vmax of the enzyme (from 7.2 to 20.5 pmol/min/mg) and induced a time- and dose-dependent phosphorylation on p70S6K residues Thr389 and Thr421/Ser424. The immunosuppressant macrolide rapamycin caused either a decrease in intensity of phospho-Thr389 bands in Western blots, or as a downshift in the relative mobility of phospho-Thr421/Ser424 bands (consistent with the loss of phosphate), but not both simultaneously. The immunosuppressant FK506 failed to inhibit p70S6K activation, but was able to rescue the rapamycin-induced downshift, pointing to a role for the mammalian target of rapamycin (mTOR) kinase. Rapamycin also caused an inhibition (IC50 0.2 nm) of the in vitro enzymatic activity of p70S6K. However, the inhibition of activity was not complete, but only a 40-50%, indicating that neutrophil p70S6K activity has a rapamycin-resistant component. This component was totally inhibited by pre-incubating the cells with the mitogen-activated protein kinase (MAPK) kinase (MEK) inhibitor PD-98059 prior to treatment with rapamycin. This indicated that a kinase from the MEK/MAPK pathway also plays a role in p70S6K activation. Thus, GM-CSF causes the dual activation of a rapamycin-resistant, MAPK-related kinase, that targets Thr421/Ser424 S6K phosphorylation, and a rapamycin-sensitive, mTOR-related kinase, that targets Thr389, both of which are needed in cooperation to achieve full activation of neutrophil p70S6K.
Collapse
Affiliation(s)
- Jason A Lehman
- Department of Physiology and Biophysics, Wright State University School of Medicine, Dayton, Ohio 45435, USA
| | | | | |
Collapse
|
13
|
Kanayasu-Toyoda T, Yamaguchi T, Oshizawa T, Uchida E, Hayakawa T. The role of c-Myc on granulocyte colony-stimulating factor-dependent neutrophilic proliferation and differentiation of HL-60 cells. Biochem Pharmacol 2003; 66:133-40. [PMID: 12818373 DOI: 10.1016/s0006-2952(03)00247-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We have previously suggested that phosphatidylinositol 3-kinase (PI3K)/p70 S6 kinase (p70 S6K) plays an important role in the regulation of neutrophilic differentiation of HL-60 cells on the basis of analysis of transferrin receptor (Trf-R)-positive (Trf-R(+)) and -negative (Trf-R(-)) cells that appear after treatment with dimethyl sulfoxide (DMSO). In the present study, we analyzed the downstream events of p70 S6K in differentiation and proliferation of both cell types, with a particular focus on c-Myc. Similar to p70 S6K, we found that the expression of c-Myc in Trf-R(+) cells is also higher than that in Trf-R(-) cells. Wortmannin, a specific inhibitor of PI3K, partially inhibited G-CSF-induced p70 S6K activity, c-Myc expression, and G-CSF-dependent proliferation, whereas rapamycin, an inhibitor of p70 S6K, completely inhibited p70 S6K activity, c-Myc expression, and G-CSF-dependent proliferation, indicating that the extent of c-Myc inhibition by these inhibitors correlates with a reduction in proliferation, and that c-Myc is downstream from PI3K/p70 S6K. We also determined phosphorylation of the 4E-binding protein 1 (4E-BP1), which is regulated downstream of the mammalian target of rapamycin. The addition of G-CSF failed to enhance the phosphorylation state of 4E-BP1 of HL-60 cells 2 days after DMSO differentiation. An antisense oligonucleotide for c-myc inhibited both G-CSF-dependent enhancement of c-Myc expression and proliferation in Trf-R(+) cells, but did not enhance the differentiation in terms of O(2)(-)-generating ability or fMLP-R expression. In contrast, antisense oligonucleotide for c-myc promoted fMLP-R on non-treated HL-60 cells. We therefore conclude that the PI3K/p70 S6K/c-Myc cascade plays an important role in neutrophilic proliferation in HL-60 cells. Unlike that of rapamycin, however, the antisense oligonucleotide for c-myc could not promote differentiation of Trf-R(+) cells cultured with G-CSF, indicating that another target downstream of p70 S6K may control the differentiation of HL-60 cells in terms of the signal transduction of G-CSF.
Collapse
Affiliation(s)
- Toshie Kanayasu-Toyoda
- Division of Cellular and Gene Therapy Products, National Institute of Health, 1-18-1, Kamiyoga, Setagaya-Ku, 158-8501 Tokyo, Japan
| | | | | | | | | |
Collapse
|
14
|
Takeda Y, Fu J, Suzuki K, Sendo D, Nitto T, Sendo F, Araki Y. Expression of GPI-80, a beta2-integrin-associated glycosylphosphatidylinositol-anchored protein, requires neutrophil differentiation with dimethyl sulfoxide in HL-60 cells. Exp Cell Res 2003; 286:199-208. [PMID: 12749849 DOI: 10.1016/s0014-4827(03)00071-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
GPI-80 is a member of the amidohydrolase family that has been proposed as a potential regulator of beta2-integrin-dependent leukocyte adhesion. GPI-80 is expressed mainly in human neutrophils. Our previous studies suggested that GPI-80 expression might be associated with myeloid differentiation. To verify this, we examined whether GPI-80 is expressed on the human promyelocytic leukemia cell line HL-60 following treatment with differentiation inducers. GPI-80 expression was induced in cells treated with dimethyl sulfoxide (DMSO) to stimulate differentiation down the neutrophil pathway. On the other hand, all-trans-retinoic acid (ATRA), another neutrophil-inducing reagent, induced no clear GPI-80 expression. Potent monocyte-inducing reagents such as 1alpha,25-dihydroxyvitamin D(3) or phorbol 12-myristate 13-acetate also had no significant effect on the protein expression. GPI-80-positive cells were found in the well-differentiated CD11b-positive and transferrin-receptor-negative cell population. Granulocyte colony-stimulating factor, which augments neutrophil differentiation of HL-60 cells, up-regulated GPI-80 expression in the presence of DMSO. Granulocyte/macrophage colony-stimulating factor, which is known to suppress the neutrophil maturation of cells, inhibited expression. Adhesion of DMSO-induced cells was regulated by anti-GPI-80 monoclonal antibody, similar to the regulation observed in neutrophils. These results suggest that use of DMSO to induce neutrophil differentiation provides suitable conditions for GPI-80 expression, and that this culture system may be a helpful model for further study of the regulation of GPI-80 expression during myeloid differentiation.
Collapse
Affiliation(s)
- Yuji Takeda
- Department of Immunology and Parasitology, Yamagata University School of Medicine, Yamagata-City, 990-9585, Japan
| | | | | | | | | | | | | |
Collapse
|
15
|
Sehgal PB, Kumar V, Guo G, Murray WC. Different patterns of regulation of Tyr-phosphorylated STAT1 and STAT3 in human hepatoma Hep3B cells by the phosphatase inhibitor orthovanadate. Arch Biochem Biophys 2003; 412:242-50. [PMID: 12667488 DOI: 10.1016/s0003-9861(03)00050-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The cellular physiology of signal transducer and activator of transcription protein family (STAT) transcription factors includes activation by Tyr-phosphorylation (PY) in cytokine and growth factor receptor complexes at the level of plasma membrane rafts, subsequent cytoplasmic transit and nuclear import, and transcriptional regulation of target genes, followed by dephosphorylation and export back to the cytoplasm. The ubiquitous protein tyrosine phosphatase (PTP) called "T-cell protein tyrosine phosphatase" has been reported to mediate Tyr-dephosphorylation of both interferon-gamma (IFN-gamma)-induced PY-STAT1 and interkleukin-6 (IL-6)-induced PY-STAT3 in some cell lines. To test whether the same PTP regulated both PY-STAT1 and PY-STAT3 in human hepatocytes we used orthovanadate (VO(4); 0.01-1.0mM) as a PTP-inhibitory probe and evaluated the kinetics of PY-STAT3 and PY-STAT1 accumulation, nuclear trafficking, and dephosphorylation following cytokine (IL-6 or IFN-gamma) stimulation of Hep3B cells. As evaluated using DNA binding or Western blotting assays, in IL-6-treated hepatocytes VO(4) had a modest enhancing effect on peak levels of cytoplasmic and nuclear PY-STAT3 reached by 1h and on their subsequent decline. In contrast, in the same cells and at the same time, VO(4) caused a marked and continuing increase in cytoplasmic and nuclear levels of PY-STAT1 which, by 4h, were 5- to 10-fold higher than peak levels reached in VO(4)-free, IL-6-treated cells. Prolonged treatment of cells with VO(4) alone (for 4-8h) replicated this markedly selective enhancement of PY-STAT1 levels. Consistent with this selectivity, shorter term VO(4) treatment (1-2h) markedly increased PY-STAT1 levels in all cellular compartments of IFN-gamma-treated cells by >10-fold. The unexpected selectivity in the effects of VO(4) on PY-STAT1 compared to that on PY-STAT3 levels in Hep3B cells suggests that, at least in these hepatocytes, the regulation of PY-STAT1 and PY-STAT3 likely involves distinct protein tyrosine phosphatase mechanisms.
Collapse
Affiliation(s)
- Pravin B Sehgal
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA.
| | | | | | | |
Collapse
|
16
|
Kanayasu-Toyoda T, Yamaguchi T, Oshizawa T, Kogi M, Uchida E, Hayakawa T. Role of the p70 S6 kinase cascade in neutrophilic differentiation and proliferation of HL-60 cells-a study of transferrin receptor-positive and -negative cells obtained from dimethyl sulfoxide- or retinoic acid-treated HL-60 cells. Arch Biochem Biophys 2002; 405:21-31. [PMID: 12176053 DOI: 10.1016/s0003-9861(02)00330-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Previously, we suggested that p70 S6 kinase (p70 S6K) plays an important role in the regulation of neutrophilic differentiation of HL-60 cells; this conclusion was based on our analysis of transferrin receptor (Trf-R) positive (Trf-R(+)) and negative (Trf-R(-)) cells that appeared after treatment with dimethyl sulfoxide (Me(2)SO). In this study, we analyzed the upstream of p70 S6K in relation to the differentiation and proliferation of both cell types. The granulocyte colony-stimulating factor (G-CSF)-induced enhancement of phosphatidylinositol 3-kinase (PI3K) activity in Trf-R(+) cells was markedly higher than that in Trf-R(-) cells. Wortmannin, a specific inhibitor of PI3K, partially inhibited G-CSF-induced p70 S6K activity and G-CSF-dependent proliferation, whereas rapamycin, an inhibitor of p70 S6K, completely inhibited these activities. The wortmannin-dependent enhancement of neutrophilic differentiation was similar to that induced by rapamycin. From these results, we conclude that the PI3K/p70 S6K cascade may play an important role in negative regulation of neutrophilic differentiation in HL-60 cells. For the G-CSF-dependent proliferation, however, p70 S6K appears to be a highly important pathway through not only a PI3K-dependent but also possibly an independent cascade.
Collapse
Affiliation(s)
- Toshie Kanayasu-Toyoda
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya-Ku, Tokyo 158-8501, Japan
| | | | | | | | | | | |
Collapse
|
17
|
Yuo A. Differentiation, apoptosis, and function of human immature and mature myeloid cells: intracellular signaling mechanism. Int J Hematol 2001; 73:438-452. [PMID: 11503957 DOI: 10.1007/bf02994005] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Human myeloid cells include hematopoietic cells at various stages of differentiation, from immature myeloid cells to mature phagocytes. Normal immature myeloid cells undergo differentiation concomitantly with proliferation in response to hematopoietic growth factors, and terminally differentiated cells, ie, mature phagocytes, exert their effector functions and then die a natural death via apoptosis. However, leukemic myeloid cells are induced to differentiate with growth suppression by several inducers, such as retinoic acid. This review describes differentiation, apoptosis, and functionality of human myeloid cells. mainly focusing on the intracellular signaling mechanism. The signal transduction system for these biological events of the life cycle of myeloid cells has recently been studied, and several characteristics have been elucidated. First, the signaling pathway for myeloid differentiation is mainly focused in the mitogen-activated protein kinases, such as extracellular signal-regulated kinase and p38, and transcriptional factors such as the signal transducers and activators of transcription PU.1 and CCAAT enhancer binding protein. Second, the signaling mechanism for myeloid cell apoptosis is fundamentally identical to that found in other cells. Caspases, caspase-activated DNase, and mitochondrial molecules such as apoptosis-inducing factor have been reported to be important, and mitogen-activated protein kinases such as p38 appear to be less important. Finally, p38 and phosphatidylinositol 3-kinase play critical roles in the signaling cascade for functional activation of mature phagocytes. The reasons why the same signaling molecules play distinct roles according to the differentiation stage and biological event await future clarification.
Collapse
Affiliation(s)
- Akira Yuo
- Department of Hematology, Research Institute, International Medical Center of Japan, Tokyo, Japan.
| |
Collapse
|
18
|
Abstract
The binding of a large number of cytokines and growth factors to their cognate receptors on the surface of mammalian-cell plasma membrane activates a signalling cascade involving the cytoplasmic STAT-family proteins, which is characterized by the nuclear translocation of a cytokine- or growth factor-specific subset of the cytoplasmic pool of the respective tyrosine- and serine-phosphorylated STAT proteins and the consequent transcriptional activation of specific target genes. In the standard model of cytokine-induced STAT signalling such as that elicited by various interferons and interleukins, it is thought that STAT proteins are recruited to the cytoplasmic side of the cell-surface receptor complex from within a monomeric cytosolic pool, and upon tyrosine-phosphorylation by respective Janus kinase family members, dimerize and translocate to the nucleus. The mechanisms which determine and regulate the recruitment of cytosolic STAT proteins to the plasma membrane-receptor complex, the transit of "activated" STATs through the expanse of the cytoplasmic compartment from the plasma membrane to the nuclear pore region, and the transit of STATs through the nuclear pore complex into the nuclear compartment, remain largely unknown. New data from different laboratories suggests consideration of a model for STAT signalling in which STAT proteins function in the cytoplasm not only as free monomers and dimers but as part of heteromeric complexes ("statosomes"), with accessory proteins which may serve to present specific STATs to the plasma membrane-receptor complex, and to chaperone "activated" STATs through the cytoplasmic compartment toward the nucleus and then into the nuclear compartment.
Collapse
Affiliation(s)
- P B Sehgal
- Departments of Cell Biology & Anatomy, and Department of Medicine, Room 201 Basic Science Building, New York Medical College, Valhalla, NY 10595, USA.
| |
Collapse
|
19
|
Calvo KR, Sykes DB, Pasillas M, Kamps MP. Hoxa9 immortalizes a granulocyte-macrophage colony-stimulating factor-dependent promyelocyte capable of biphenotypic differentiation to neutrophils or macrophages, independent of enforced meis expression. Mol Cell Biol 2000; 20:3274-85. [PMID: 10757811 PMCID: PMC85621 DOI: 10.1128/mcb.20.9.3274-3285.2000] [Citation(s) in RCA: 114] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The genes encoding Hoxa9 and Meis1 are transcriptionally coactivated in a subset of acute myeloid leukemia (AML) in mice. In marrow reconstitution experiments, coexpression of both genes produces rapid AML, while neither gene alone generates overt leukemia. Although Hoxa9 and Meis1 can bind DNA as heterodimers, both can also heterodimerize with Pbx proteins. Thus, while their coactivation may result from the necessity to bind promoters as heterodimers, it may also result from the necessity of altering independent biochemical pathways that cooperate to generate AML, either as monomers or as heterodimers with Pbx proteins. Here we demonstrate that constitutive expression of Hoxa9 in primary murine marrow immortalizes a late myelomonocytic progenitor, preventing it from executing terminal differentiation to granulocytes or monocytes in the presence of granulocyte-macrophage colony-stimulating factor (GM-CSF) or interleukin-3. This immortalized phenotype is achieved in the absence of endogenous or exogenous Meis gene expression. The Hoxa9-immortalized progenitor exhibited a promyelocytic transcriptional profile, expressing PU.1, AML1, c-Myb, C/EBP alpha, and C/EBP epsilon as well as their target genes, the receptors for GM-CSF, G-CSF, and M-CSF and the primary granule proteins myeloperoxidase and neutrophil elastase. G-CSF obviated the differentiation block of Hoxa9, inducing neutrophilic differentiation with accompanying expression of neutrophil gelatinase B and upregulation of gp91phox. M-CSF also obviated the differentiation block, inducing monocytic differentiation with accompanying expression of the macrophage acetyl-low-density lipoprotein scavenger receptor and F4/80 antigen. Versions of Hoxa9 lacking the ANWL Pbx interaction motif (PIM) also immortalized a promyelocytic progenitor with intrinsic biphenotypic differentiation potential. Therefore, Hoxa9 evokes a cytokine-selective block in differentiation by a mechanism that does not require Meis gene expression or interaction with Pbx through the PIM.
Collapse
Affiliation(s)
- K R Calvo
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, California 92093-0612, USA.
| | | | | | | |
Collapse
|
20
|
Wen X, Lin HH, Shih HM, Kung HJ, Ann DK. Kinase activation of the non-receptor tyrosine kinase Etk/BMX alone is sufficient to transactivate STAT-mediated gene expression in salivary and lung epithelial cells. J Biol Chem 1999; 274:38204-10. [PMID: 10608894 DOI: 10.1074/jbc.274.53.38204] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Etk/BMX is a non-receptor protein tyrosine kinase that requires a functional phosphatidylinositol 3-kinase via the pleckstrin homology domain to be activated by cytokine. In the present study, a conditionally active form of Etk was constructed by fusing the hormone-binding domain of estrogen receptor (ER) to an amino terminus truncated form of Etk, PHDelta1-68Etk, to generate DeltaEtk:ER. In stably transfected Pa-4DeltaEtk:ER cells, the activity of DeltaEtk:ER was stimulated within minutes by the treatment of DeltaEtk:ER stimulant, estradiol, and sustained for greater than 24 h. A robust induction in the phosphorylation of signal transducers and activators of transcription (STAT) proteins, including STAT1, STAT3, and STAT5, was accompanied with DeltaEtk:ER activation. Moreover, the conditionally activated Etk stimulated STAT1- and STAT5-dependent reporter activities by approximately 160- and approximately 15-fold, respectively, however, elicited only a modest STAT3-mediated reporter activation. Qualitatively comparable results were obtained in lung A549 cells, indicating that DeltaEtk:ER inducible system could function in an analogous fashion in different epithelial cells. Furthermore, we demonstrated that Etk activation alone augmented cyclin D1 promoter/enhancer activity via its STAT5 response element in both Pa-4DeltaEtk:ER and A549 cells. Altogether, these findings support the notion that the activation of Etk kinase is sufficient to transactivate STAT-mediated gene expression. Hence, our inducible DeltaEtk:ER system represents a novel approach to investigate the biochemical events following Etk activation and to evaluate the contribution by kinase activation of Etk alone or in conjunction with other signaling pathway(s) to the ultimate biological responses.
Collapse
Affiliation(s)
- X Wen
- Department of Molecular Pharmacology, University of Southern California, Los Angeles, California 90033, USA
| | | | | | | | | |
Collapse
|