1
|
Hu J, Zheng L, Shen X, Zhang Y, Li C, Xi T. MicroRNA-125b inhibits AML cells differentiation by directly targeting Fes. Gene 2017; 620:1-9. [PMID: 28389358 DOI: 10.1016/j.gene.2017.04.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 02/09/2017] [Accepted: 04/03/2017] [Indexed: 01/05/2023]
Abstract
MicroRNA-125b (miR-125b) has been reported to be upregulated in several kinds of leukemia, suggesting that miR-125b plays a role in Leukemia development. In this study, it was shown that miR-125b expression level decreased in response to 1α, 25-dihydroxy-vitamin D3 (1,25D3) in a dose- and time-dependent manner and miR-125b blocked 1,25D3-induced monocytic differentiation of U937 cells. In addition, miR-125b decreased mRNA expression of myelomonocytic differentiation markers, including CD11c, CD18 and CD64 and arrested the cell cycle at the S phase in U937 and HL60 cells. Fes was identified as a novel direct target of miR-125b and miR-125b could also reduce the expression levels of PU.1 and macrophage colony-stimulating factor receptor (MCSFR). Furthermore, Fes was found to be involved in monocytic differentiation via upregulation of PU.1 and MCSFR and Fes siRNA could also inhibit 1,25D3-induced monocytic differentiation of U937 and HL60 cells and decrease mRNA expression of CD11c, CD18 and CD64. Importantly, the inhibition of Fes siRNA on 1,25D3-induced monocytic differentiation could be rescued by transfection with miR-125b inhibitor. Our data highlights an important role of miR-125b in AML progression, implying the potential application of miR-125b in AML therapy.
Collapse
Affiliation(s)
- Jinhang Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Lufeng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Xiao Shen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yan Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Cheng Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Tao Xi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, People's Republic of China; Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
2
|
Wang Y, Li J, Li Y, Fang L, Sun X, Chang S, Zhao P, Cui Z. Identification of ALV-J associated acutely transforming virus Fu-J carrying complete v-fps oncogene. Virus Genes 2016; 52:365-71. [PMID: 27108997 DOI: 10.1007/s11262-016-1301-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 01/27/2016] [Indexed: 11/30/2022]
Abstract
Transduction of oncogenes by ALVs and generation of acute transforming viruses is common in natural viral infections. In order to understand the molecular basis for the rapid oncogenicity of Fu-J, an acutely transforming avian leukosis virus isolated from fibrosarcomas in crossbreed broilers infected with subgroup J avian leukosis virus (ALV-J) in China, complete genomic structure of Fu-J virus was determined by PCR amplification and compared with those of Fu-J1, Fu-J2, Fu-J3, Fu-J4, and Fu-J5 reported previously. The results showed that the genome of Fu-J was defective, with parts of gag gene replaced by the complete v-fps oncogene and encoded a 137 kDa Gag-fps fusion protein. Sequence analysis revealed that Fu-J and Fu-J1 to Fu-J5 were related quasi-species variants carrying different lengths of v-fps oncogenes generated from recombination between helper virus and c-fps gene. Comparison of virus carrying v-fps oncogene also gave us a glimpse of the molecular characterization and evolution process of the acutely transforming ALV.
Collapse
Affiliation(s)
- Yixin Wang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Road No. 61, Tai'an, 271018, Shandong, China
| | - Jianliang Li
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Road No. 61, Tai'an, 271018, Shandong, China
| | - Yang Li
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Road No. 61, Tai'an, 271018, Shandong, China
| | - Lichun Fang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Road No. 61, Tai'an, 271018, Shandong, China
| | - Xiaolong Sun
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Road No. 61, Tai'an, 271018, Shandong, China
| | - Shuang Chang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Road No. 61, Tai'an, 271018, Shandong, China
| | - Peng Zhao
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Road No. 61, Tai'an, 271018, Shandong, China.
| | - Zhizhong Cui
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Daizong Road No. 61, Tai'an, 271018, Shandong, China.
| |
Collapse
|
3
|
Kumar V, Everingham S, Hall C, Greer PA, Craig AWB. Calpains promote neutrophil recruitment and bacterial clearance in an acute bacterial peritonitis model. Eur J Immunol 2013; 44:831-41. [PMID: 24375267 DOI: 10.1002/eji.201343757] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 10/15/2013] [Accepted: 11/29/2013] [Indexed: 12/20/2022]
Abstract
Activation of the innate immune system is critical for clearance of bacterial pathogens to limit systemic infections and host tissue damage. Here, we report a key role for calpain proteases in bacterial clearance in mice with acute peritonitis. Using transgenic mice expressing Cre recombinase primarily in innate immune cells (fes-Cre), we generated conditional capns1 knockout mice. Consistent with capns1 being essential for stability and function of the ubiquitous calpains (calpain-1, calpain-2), peritoneal cells from these mice had reduced levels of calpain-2/capns1, and reduced proteolysis of their substrate selenoprotein K. Using an acute bacterial peritonitis model, we observed impaired bacterial killing within the peritoneum and development of bacteremia in calpain knockout mice. These defects correlated with significant reductions in IL-1α release, neutrophil recruitment, and generation of reactive oxygen species in calpain knockout mice with acute bacterial peritonitis. Peritoneal macrophages from calpain knockout mice infected with enterobacteria ex vivo, were competent in phagocytosis of bacteria, but showed impaired clearance of intracellular bacteria compared with control macrophages. Together, these results implicate calpains as key mediators of effective innate immune responses to acute bacterial infections, to prevent systemic dissemination of bacteria that can lead to sepsis.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | | | | | | |
Collapse
|
4
|
Sangrar W, Zirgnibl RA, Gao Y, Muller WJ, Jia Z, Greer PA. An Identity Crisis for fps/fes: Oncogene or Tumor Suppressor? Cancer Res 2005; 65:3518-22. [PMID: 15867340 DOI: 10.1158/0008-5472.can-04-3468] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Fps/Fes proteins were among the first members of the protein tyrosine kinase family to be characterized as dominant-acting oncoproteins. Addition of retroviral GAG sequences or other experimentally induced mutations activated the latent transforming potential of Fps/Fes. However, activating mutations in fps/fes had not been found in human tumors until recently, when mutational analysis of a panel of colorectal cancers identified four somatic mutations in sequences encoding the Fps/Fes kinase domain. Here, we report biochemical and theoretical structural analysis demonstrating that three of these mutations result in inactivation, not activation, of Fps/Fes, whereas the fourth mutation compromised in vivo activity. These results did not concur with a classic dominant-acting oncogenic role for fps/fes involving activating somatic mutations but instead raised the possibility that inactivating fps/fes mutations might promote tumor progression in vivo. Consistent with this, we observed that tumor onset in a mouse model of breast epithelial cancer occurred earlier in mice targeted with either null or kinase-inactivating fps/fes mutations. Furthermore, a fps/fes transgene restored normal tumor onset kinetics in targeted fps/fes null mice. These data suggest a novel and unexpected tumor suppressor role for Fps/Fes in epithelial cells.
Collapse
Affiliation(s)
- Waheed Sangrar
- Division of Cancer Biology and Genetics, Queen's University Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
5
|
Sangrar W, Gao Y, Bates B, Zirngibl R, Greer PA. Activated Fps/Fes tyrosine kinase regulates erythroid differentiation and survival. Exp Hematol 2004; 32:935-45. [PMID: 15504549 DOI: 10.1016/j.exphem.2004.07.004] [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] [Received: 03/09/2004] [Revised: 06/17/2004] [Accepted: 07/17/2004] [Indexed: 10/26/2022]
Abstract
OBJECTIVE A substantial body of evidence implicates the cytoplasmic protein tyrosine kinase Fps/Fes in regulation of myeloid differentiation and survival. In this study we wished to determine if Fps/Fes also plays a role in the regulation of erythropoiesis. METHODS Mice tissue-specifically expressing a "gain-of-function" mutant fps/fes transgene (fps(MF)) encoding an activated variant of Fps/Fes (MFps), were used to explore the in vivo biological role of Fps/Fes. Erythropoiesis in these mice was assessed by hematological analysis, lineage marker analysis, bone-marrow colony assays, and biochemical approaches. RESULTS fps(MF) mice displayed reductions in peripheral red cell counts. However, there was an accumulation of immature erythroid precursors, which displayed increased survival. Fps/Fes and the related Fer kinase were both detected in early erythroid progenitors/blasts and in mature red cells. Fps/Fes was also activated in response to erythropoietin (EPO) and stem cell factor (SCF), two critical factors in erythroid development. In addition, increased Stat5A/B activation and reduced Erk1/2 phosphorylation was observed in fps(MF) primary erythroid cells in response to EPO or SCF, respectively. CONCLUSIONS These data support a role for Fps/Fes in regulating the survival and differentiation of erythroid cells through modulation of Stat5A/B and Erk kinase pathways induced by EPO and SCF. The increased numbers and survival of erythroid progenitors from fps(MF) mice, and their differential responsiveness to SCF and EPO, implicates Fps/Fes in the commitment of multilineage progenitors to the erythroid lineage. The anemic phenotype in fps(MF) mice suggests that downregulation of Fps/Fes activity might be required for terminal erythroid differentiation.
Collapse
Affiliation(s)
- Waheed Sangrar
- Division of Cancer Biology and Genetics, Queen's University Cancer Research Institute, Queen's University, Kingston, Ontario, Canada
| | | | | | | | | |
Collapse
|
6
|
Sangrar W, Mewburn JD, Vincent SG, Fisher JT, Greer PA. Vascular defects in gain-of-function fps/fes transgenic mice correlate with PDGF- and VEGF-induced activation of mutant Fps/Fes kinase in endothelial cells. J Thromb Haemost 2004; 2:820-32. [PMID: 15099290 DOI: 10.1111/j.1538-7836.2004.00654.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Fps/Fes is a cytoplasmic tyrosine kinase that is abundantly expressed in the myeloid, endothelial, epithelial, neuronal and platelet lineages. Genetic manipulation in mice has uncovered potential roles for this kinase in hematopoiesis, innate immunity, inflammation and angiogenesis. OBJECTIVE We have utilized a genetic approach to explore the role of Fps/Fes in angiogenesis. METHODS A hypervascular line of mice generated by expression of a 'gain-of-function' human fps/fes transgene (fps(MF)) encoding a myristoylated variant of Fps (MFps) was used in these studies. The hypervascular phenotype of this line was extensively characterized by intravital microscopy and biochemical approaches. RESULTS fps(MF) mice exhibited 1.6-1.7-fold increases in vascularity which was attributable to increases in the number of secondary vessels. Vessels were larger, exhibited varicosities and disorganized patterning, and were found to have defects in histamine-induced permeability. Biochemical characterization of endothelial cell (EC) lines derived from fps(MF) mice revealed that MFps was hypersensitive to activation by vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF). CONCLUSIONS MFps mediates enhanced sensitization to VEGF and PDGF signaling in ECs. We propose that this hypersensitization contributes to excessive angiogenic signaling and that this underlies the observed hypervascular phenotype of fps(MF) mice. These phenotypes recapitulate important aspects of the vascular defects observed in both VEGF and angiopoietin-1 transgenic mice. The fps/fes proto-oncogene product therefore represents a novel player in the regulation of angiogenesis, and the fps(MF) line of mice constitutes a unique new murine model for the study of this process.
Collapse
Affiliation(s)
- W Sangrar
- Division of Cancer Biology and Genetics, Queen's University Cancer Research Institute, Ontario, Canada
| | | | | | | | | |
Collapse
|
7
|
Sangrar W, Gao Y, Zirngibl RA, Scott ML, Greer PA. The fps/fes proto-oncogene regulates hematopoietic lineage output. Exp Hematol 2003; 31:1259-67. [PMID: 14662333 DOI: 10.1016/j.exphem.2003.09.013] [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
OBJECTIVE The fps/fes proto-oncogene is abundantly expressed in myeloid cells, and the Fps/Fes cytoplasmic protein-tyrosine kinase is implicated in signaling downstream from hematopoietic cytokines, including interleukin-3 (IL-3), granulocyte-macrophage colony-stimulating factor (GM-CSF), and erythropoietin (EPO). Studies using leukemic cell lines have previously suggested that Fps/Fes contributes to granulomonocytic differentiation, and that it might play a more selective role in promoting survival and differentiation along the monocytic pathway. In this study we have used a genetic approach to explore the role of Fps/Fes in hematopoiesis. METHODS We used transgenic mice that tissue-specifically express a mutant human fps/fes transgene (fps(MF)) that was engineered to encode Fps/Fes kinase that is activated through N-terminal myristoylation (MFps). Hematopoietic function was assessed using lineage analysis, hematopoietic progenitor cell colony-forming assays, and biochemical approaches. RESULTS fps(MF) transgenic mice displayed a skewed hematopoietic output reflected by increased numbers of circulating granulocytic and monocytic cells and a corresponding decrease in lymphoid cells. Bone marrow colony assays of progenitor cells revealed a significant increase in the number of both granulomonocytic and multi-lineage progenitors. A molecular analysis of signaling in mature monocytic cells showed that MFps promoted GM-CSF-induced STAT3, STAT5, and ERK1/2 activation. CONCLUSIONS These observations support a role for Fps/Fes in signaling pathways that contribute to lineage determination at the level of multi-lineage hematopoietic progenitors as well as the more committed granulomonocytic progenitors.
Collapse
Affiliation(s)
- Waheed Sangrar
- Division of Cancer Biology and Genetics, Queen's University Cancer Research Institute, Kingston, Ontario, K7L 3N6, Canada
| | | | | | | | | |
Collapse
|
8
|
Senis YA, Craig AWB, Greer PA. Fps/Fes and Fer protein-tyrosinekinases play redundant roles in regulating hematopoiesis. Exp Hematol 2003; 31:673-81. [PMID: 12901971 DOI: 10.1016/s0301-472x(03)00107-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE The highly related protein-tyrosine kinases Fps (also called Fes) and Fer are sole members of a subfamily of kinases. In this study, knock-in mice harboring kinase-inactivating mutations in both fps and fer alleles were used to assess functional redundancy between Fps and Fer kinases in regulating hematopoiesis. METHODS Mice harboring kinase-inactivating mutations in fps and fer alleles were generated previously. Compound homozygous mice were bred that lack both Fps and Fer kinase activities and progeny were analyzed for potential defects in viability and fertility. Potential differences in hematopoiesis were analyzed by lineage analysis of bone marrow cells, peripheral blood counts, and hematopoietic progenitor cell colony-forming assays. RESULTS Mice devoid of both Fps and Fer kinase activities were viable and displayed reduced fertility. Circulating levels of neutrophils, erythrocytes, and platelets were elevated in compound mutant mice compared to wild-type controls, suggesting that hematopoiesis is deregulated in the absence of Fps and Fer kinases. Compound mutant mice also showed reduced overall bone marrow cellularity, and lineage analysis revealed elevated CD11b(hi)Ly-6G(lo) myeloid cells, which may reflect increased granulocyte progenitors. Although no differences in the overall number of granulocyte/monocyte colony-forming progenitors were observed, qualitative differences in myeloid colonies from compound mutant mice suggested a role for Fps and Fer kinases in regulating cell-cell adhesion or a skewing in cellularity of colonies. CONCLUSIONS Mice lacking both Fps and Fer kinase activities develop normally, show reduced fertility, and display defects in hematopoiesis, thus providing evidence for functional redundancy between Fps and Fer kinases in regulating hematopoiesis.
Collapse
Affiliation(s)
- Yotis A Senis
- Department of Pathology, Queen's University, Kingston, Ontario, Canada
| | | | | |
Collapse
|
9
|
Greaves DR, Gordon S. Macrophage-specific gene expression: current paradigms and future challenges. Int J Hematol 2002; 76:6-15. [PMID: 12138897 DOI: 10.1007/bf02982713] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cells of the mononuclear phagocyte lineage include macrophages, microglia, osteoclasts, and myeloid dendritic cells. These cell types are all derived from blood monocytes, which are the product of hematopoietic stem cell differentiation. In this review we use specific examples of macrophage-expressed genes to illustrate potential regulatory strategies for directing macrophage-specific gene expression. The examples we have chosen-the human c-fes gene, the murine spi-1 (PU.1) gene, the human RANTES promoter, and the human CD68 gene-illustrate different aspects of constitutive and inducible gene expression in macrophages. One important challenge for future work in this field will be to identify the molecular events that dictate lineage decisions during the differentiation of mononuclear phagocytes from hematopoietic progenitor cells. Another important goal will be to understand how groups of macrophage genes are coordinately expressed in response to physiological, immunological, and inflammatory stimuli. A better understanding of macrophage gene expression may find application in gene therapy, genetic vaccination, and the development of new antiinflammatory drugs.
Collapse
Affiliation(s)
- David R Greaves
- Sir William Dunn School of Pathology, University of Oxford, United Kingdom.
| | | |
Collapse
|
10
|
Abstract
Fps/Fes and Fer are the only known members of a distinct subfamily of the non-receptor protein-tyrosine kinase family. Recent studies indicate that these kinases have roles in regulating cytoskeletal rearrangements and inside out signalling that accompany receptor ligand, cell matrix and cell cell interactions. Genetic analysis using transgenic mouse models also implicates these kinases in the regulation of inflammation and innate immunity.
Collapse
MESH Headings
- Animals
- Biological Evolution
- Chromosomes, Human, Pair 15/genetics
- Chromosomes, Human, Pair 5/genetics
- Fusion Proteins, gag-onc/chemistry
- Fusion Proteins, gag-onc/genetics
- Fusion Proteins, gag-onc/physiology
- Humans
- Inflammation/physiopathology
- Mice
- Mice, Knockout
- Mice, Transgenic
- Models, Biological
- Models, Molecular
- Protein Structure, Tertiary
- Protein-Tyrosine Kinases/chemistry
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/physiology
- Proto-Oncogene Proteins/chemistry
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/physiology
- Receptor Cross-Talk
- Receptors, Platelet-Derived Growth Factor/physiology
- Signal Transduction
Collapse
Affiliation(s)
- Peter Greer
- Division of Cancer Research and Genetics, Queen's University Cancer Research Institute, Kingston, Ontario K7L 3N6, Canada.
| |
Collapse
|
11
|
Abstract
Myeloid blood cells comprise an important component of the immune system. Proper control of both lineage- and stage-specific gene expression is required for normal myeloid cell development and function. In recent years, a relatively small number of critical transcriptional regulators have been identified that serve important roles both in myeloid cell development and regulation of lineage-restricted gene expression in mature myeloid cells. This review summarizes our current understanding of the regulation of lineage- and stage-restricted transcription during myeloid cell differentiation, how critical transcriptional regulators control myeloid cell development, and how perturbations in transcription factor function results in the development of leukemia.
Collapse
Affiliation(s)
- David G Skalnik
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
12
|
Keller P, Payne JL, Tremml G, Greer PA, Gaboli M, Pandolfi PP, Bessler M. FES-Cre targets phosphatidylinositol glycan class A (PIGA) inactivation to hematopoietic stem cells in the bone marrow. J Exp Med 2001; 194:581-9. [PMID: 11535627 PMCID: PMC2195941 DOI: 10.1084/jem.194.5.581] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A somatic mutation in the X-linked phosphatidylinositol glycan class A (PIGA) gene causes the loss of glycosyl phosphatidylinositol (GPI)-linked proteins on blood cells from patients with paroxysmal nocturnal hemoglobinuria. Because all blood cell lineages may be affected it is thought that the mutation occurs in a hematopoietic stem cell. In transgenic mice, germline transmission of an inactive Piga gene is embryonic lethal. To inactivate the murine Piga gene in early hematopoiesis we therefore chose conditional gene inactivation using the Cre/loxP system. We expressed Cre recombinase under the transcription regulatory sequences of the human c-fes gene. FES-Cre inactivated PIGA in hematopoietic cells of mice carrying a floxed Piga allele (LF mice). PIGA(-) cells were found in all hematopoietic lineages of definitive but not primitive hematopoiesis. Their proportions were low in newborn mice but subsequently increased continuously to produce for the first time mice that have almost exclusively PIGA(-) blood cells. The loss of GPI-linked proteins occurred mainly in c-kit(+)CD34(+)Lin(-) progenitor cells before the CFU-GEMM stage. Using bone marrow reconstitution experiments with purified PIGA(-) cells we demonstrate that LF mice have long-term bone marrow repopulating cells that lack GPI-linked proteins, indicating that recombination of the floxed Piga allele occurs in the hematopoietic stem cell.
Collapse
Affiliation(s)
- Peter Keller
- Division of Hematology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Jennifer L. Payne
- Division of Hematology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Gabi Tremml
- Department of Human Genetics, Memorial Sloan-Kettering Cancer Center, New York, NY 10021
| | - Peter A. Greer
- Cancer Research Laboratories, Departments of Biochemistry and Pathology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Mirella Gaboli
- Department of Human Genetics, Memorial Sloan-Kettering Cancer Center, New York, NY 10021
| | - Pier P. Pandolfi
- Department of Human Genetics, Memorial Sloan-Kettering Cancer Center, New York, NY 10021
| | - Monica Bessler
- Division of Hematology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
13
|
Jiang H, Foltenyi K, Kashiwada M, Donahue L, Vuong B, Hehn B, Rothman P. Fes mediates the IL-4 activation of insulin receptor substrate-2 and cellular proliferation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:2627-34. [PMID: 11160325 DOI: 10.4049/jimmunol.166.4.2627] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although Jak kinases are essential for initiating cytokine signaling, the role of other nonreceptor tyrosine kinases in this process remains unclear. We have examined the role of Fes in IL-4 signaling. Examination of Jak1-deficient cell lines demonstrates that Jak1 is required for the activation of Fes by IL-4. Experiments studying signaling molecules activated by IL-4 receptor suggest that IL-4 signaling can be subdivided into Fes-dependent and Fes-independent pathways. Overexpression of kinase-inactive Fes blocks the IL-4 activation of insulin receptor substrate-2, but not STAT6. Fes appears to be a downstream kinase from Jak1/Jak3 in this process. Further examination of downstream signaling demonstrates that kinase-inactive Fes inhibits the recruitment of phosphoinositide 3-kinase to the activated IL-4 receptor complex and decreases the activation of p70(S6k) kinase in response to IL-4. This inhibition correlates with a decrease in IL-4-induced proliferation. In contrast, mutant Fes does not inhibit the activation of Akt by IL-4. These data demonstrate that signaling pathways activated by IL-4 require different tyrosine kinases. This differential requirement predicts that specific kinase inhibitors may permit the disruption of specific IL-4-induced functions.
Collapse
Affiliation(s)
- H Jiang
- Department of Medicine and Microbiology, Columbia University, College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
AbstractThe c-fes proto-oncogene encodes a 92-kd protein tyrosine kinase whose expression is restricted largely to myeloid and endothelial cells in adult mammals. A 13.2-kilobase (kb) humanc-fes genomic fragment was previously shown to containcis-acting element(s) sufficient for a locus control function in bone marrow macrophages. Locus control regions (LCRs) confer transgene expression in mice that is integration site independent, copy number dependent, and similar to endogenous murine messenger RNA levels. To identify sequences required for this LCR,c-fes transgenes were analyzed in mice. Myeloid-cell–specific, deoxyribonuclease-I–hypersensitive sites localized to the 3′ boundary of exon 1 and intron 3 are required to confer high-level transgene expression comparable to endogenous c-fes, independent of integration site. We define a minimal LCR element as DNA sequences (nucleotides +28 to +2523 relative to the transcription start site) located within intron 1 to intron 3 of the human locus. When this 2.5-kb DNA fragment was linked to a c-fes complementary DNA regulated by its own 446–base-pair promoter, integration-site–independent, copy-number–dependent transcription was observed in myeloid cells in transgenic mice. Furthermore, this 2.5-kb cassette directed expression of a heterologous gene (enhanced green fluorescent protein) exclusively in myeloid cells. The c-fes regulatory unit represents a novel reagent for targeting gene expression to macrophages and neutrophils in transgenic mice.
Collapse
|
15
|
A minimal c-fes cassette directs myeloid-specific expression in transgenic mice. Blood 2000. [DOI: 10.1182/blood.v96.9.3040.h8003040_3040_3048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The c-fes proto-oncogene encodes a 92-kd protein tyrosine kinase whose expression is restricted largely to myeloid and endothelial cells in adult mammals. A 13.2-kilobase (kb) humanc-fes genomic fragment was previously shown to containcis-acting element(s) sufficient for a locus control function in bone marrow macrophages. Locus control regions (LCRs) confer transgene expression in mice that is integration site independent, copy number dependent, and similar to endogenous murine messenger RNA levels. To identify sequences required for this LCR,c-fes transgenes were analyzed in mice. Myeloid-cell–specific, deoxyribonuclease-I–hypersensitive sites localized to the 3′ boundary of exon 1 and intron 3 are required to confer high-level transgene expression comparable to endogenous c-fes, independent of integration site. We define a minimal LCR element as DNA sequences (nucleotides +28 to +2523 relative to the transcription start site) located within intron 1 to intron 3 of the human locus. When this 2.5-kb DNA fragment was linked to a c-fes complementary DNA regulated by its own 446–base-pair promoter, integration-site–independent, copy-number–dependent transcription was observed in myeloid cells in transgenic mice. Furthermore, this 2.5-kb cassette directed expression of a heterologous gene (enhanced green fluorescent protein) exclusively in myeloid cells. The c-fes regulatory unit represents a novel reagent for targeting gene expression to macrophages and neutrophils in transgenic mice.
Collapse
|
16
|
Largaespada DA. Genetic heterogeneity in acute myeloid leukemia: maximizing information flow from MuLV mutagenesis studies. Leukemia 2000; 14:1174-84. [PMID: 10914539 DOI: 10.1038/sj.leu.2401852] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The study of myeloid leukemia induced by slow transforming murine leukemia viruses (MuLV) in the laboratory mouse has led to discovery of many important genes with critical roles in regulating the growth, death, lineage determination and development of hematopoietic precursor cells. This review provides an overview of the susceptible strains and virus isolates that cause acute myeloid leukemia (AML) in mice. In addition, newer methodologies, involving the use of the polymerase chain reaction, that have been used to identify cancer genes mutated by proviral insertion in mouse models, will be discussed. As cancer is a multi-gene disease, a system in which pairs of oncogenic mutations are classified as redundant, neutral or synergistic is described. The potential to combine MuLV mutagenesis with recent advances in mouse transgenesis in order to model specific forms of myeloid leukemia or genetic pathways common in human AML will be discussed. Finally, a general strategy for maximizing these genetically rich models to foster a better understanding of AML physiology and developing therapies is proposed.
Collapse
MESH Headings
- Acute Disease
- Animals
- Biological Specimen Banks
- Crosses, Genetic
- DNA, Neoplasm/genetics
- Databases, Factual
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic
- Gene Library
- Genetic Heterogeneity
- Genetic Predisposition to Disease
- Humans
- Leukemia Virus, Murine/genetics
- Leukemia Virus, Murine/isolation & purification
- Leukemia Virus, Murine/pathogenicity
- Leukemia, Experimental/genetics
- Leukemia, Experimental/pathology
- Leukemia, Experimental/virology
- Leukemia, Myeloid/genetics
- Leukemia, Myeloid/pathology
- Leukemia, Myeloid/virology
- Mice
- Mice, Inbred Strains/genetics
- Mice, Inbred Strains/virology
- Mice, Transgenic
- Mutagenesis, Insertional
- Oncogenes
- Polymerase Chain Reaction
- Proto-Oncogenes
- Proviruses/genetics
- Retroviridae Infections/genetics
- Retroviridae Infections/pathology
- Retroviridae Infections/virology
- Transgenes
- Tumor Virus Infections/genetics
- Tumor Virus Infections/pathology
- Tumor Virus Infections/virology
Collapse
Affiliation(s)
- D A Largaespada
- Department of Genetics, Cell Biology and Development, Institute of Human Genetics, and University of Minnesota Cancer Center, University of Minnesota, Minneapolis 55455, USA
| |
Collapse
|
17
|
Kim L, Wong TW. Growth factor-dependent phosphorylation of the actin-binding protein cortactin is mediated by the cytoplasmic tyrosine kinase FER. J Biol Chem 1998; 273:23542-8. [PMID: 9722593 DOI: 10.1074/jbc.273.36.23542] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previous characterization of the nonreceptor tyrosine kinase FER identified a tight physical association with the catenin pp120 and led to the suggestion that FER may be involved in cell-cell signaling. To further understand the function of FER, we have continued our analyses of the interaction of FER with pp120 and other proteins. The majority of FER is localized to the cytoplasmic fraction where it forms a complex with the actin-binding protein cortactin. The Src homology 2 sequence of FER is required for directly binding cortactin, and phosphorylation of the FER-cortactin complex is up-regulated in cells treated with peptide growth factors. Using a dominant-negative mutant of FER, we provided evidence that FER kinase activity is required for the growth factor-dependent phosphorylation of cortactin. These data suggest that cortactin is likely to be a direct substrate of FER. Our observations provide additional support for a role of FER in mediating signaling from the cell surface, via growth factor receptors, to the cytoskeleton. The nature of the FER-cortactin interaction, and their putative enzyme-substrate relationship, support the previous proposal that one of the functions of the Src homology 2 sequences of nonreceptor tyrosine kinases is to provide a binding site for their preferred substrates.
Collapse
Affiliation(s)
- L Kim
- Department of Biochemistry, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA
| | | |
Collapse
|
18
|
Schwartz Y, Ben-Dor I, Navon A, Motro B, Nir U. Tyrosine phosphorylation of the TATA element modulatory factor by the FER nuclear tyrosine kinases. FEBS Lett 1998; 434:339-45. [PMID: 9742951 DOI: 10.1016/s0014-5793(98)01003-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The FER locus in the mouse encodes two tyrosine kinases, p94fer and p51ferT. While p94fer accumulates in the cytoplasm and nucleus of most mammalian cells the expression of p51ferT is restricted to the nucleus of meiotic primary spermatocytes. The cellular function of the FER kinases is not understood, nor has a substrate for these enzymes been characterized. To identify putative substrates of p94fer and p51ferT, the two enzymes were used as 'baits' in the yeast two-hybrid screening system. cDNAs encoding the mouse TATA element modulatory factor (TMF) were repeatedly isolated in this assay. TMF was previously shown to bind the TATA element in RNA polymerase II promoters and impaired their functioning in a cell free transcription system. Both p94fer and p51ferT phosphorylated the TMF protein in in vitro and in vivo kinase assays. Sequential deletions showed that the carboxy-terminal region of TMF was essential for phosphorylation. In situ hybridization analysis revealed the preferential accumulation of TMF transcripts in meiotic spermatogenic and oogenic cells. p94fer and p51ferT may thus modulate the suppressive activity of TMF during cellular growth and in defined differentiation processes.
Collapse
Affiliation(s)
- Y Schwartz
- Department of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | | | | | | |
Collapse
|
19
|
Heydemann A, Boehmler JH, Simon MC. Expression of two myeloid cell-specific genes requires the novel transcription factor, c-fes expression factor. J Biol Chem 1997; 272:29527-37. [PMID: 9368014 DOI: 10.1074/jbc.272.47.29527] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The protein product of the c-fes proto-oncogene has been implicated in the normal development of myeloid cells (macrophages and granulocytes). We have previously shown that 151 base pairs of c-fes 5'-flanking sequences are sufficient for myeloid cell-specific expression and include functional binding sites for Sp1, PU.1, and a novel nuclear factor (Heydemann, A., Juang, G., Hennessy, K., Parmacek, M. S., and Simon, M. C. (1996) Mol. Cell. Biol. 16, 1676-1686). This novel hematopoietic transcription factor, termed FEF (c-fes expression factor), binds to a cis-acting element that is located at nucleotides -9 to -4 of the c-fes promoter between two Ets binding sites (at -19 to -15 and -4 to +1) which bind PU.1. We now show that a FEF binding site exists in the myeloid cell-specific regulatory region of a second gene, the -2.7-kilobase pair enhancer of chicken lysozyme. The lysozyme FEF site is immediately 5' to a PU. 1 site, analogous to their arrangement in the c-fes promoter, and allows the formation of a preliminary FEF consensus site, 5'-GAAT(C/G)A-3'. This consensus site does not match any sites for known transcription factors. Importantly, although PU.1 binds immediately 3' of the FEF site in both the c-fes promoter and the chicken lysozyme enhancer (CLE), we show that they bind independently. The FEF sites are required for high levels of transcription by both the CLE and the c-fes promoter in transient transfection experiments. Importantly, elimination of the CLE FEF site abolishes all transcriptional activity of this enhancer element. Mutation of the adjacent PU.1 site in either the c-fes promoter or the CLE, reduces activity by approximately 50%. Therefore, transcription of both lysozyme and fes in myeloid cells requires FEF and PU.1. UV cross-linking experiments show that the FEF binding activity consists of a single 70-kDa protein in both human and murine cell lines. FEF binding activity is not affected by antibodies that specifically recognize a number of cloned transcription factors. Collectively, these data indicate that we have identified a novel transcription factor that is functionally important for the expression of at least two myeloid cell-specific genes.
Collapse
Affiliation(s)
- A Heydemann
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois 60637, USA
| | | | | |
Collapse
|
20
|
|
21
|
|
22
|
Sturrock A, Franklin KF, Hoidal JR. Human proteinase-3 expression is regulated by PU.1 in conjunction with a cytidine-rich element. J Biol Chem 1996; 271:32392-402. [PMID: 8943304 DOI: 10.1074/jbc.271.50.32392] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Human proteinase-3 is one of three serine proteinases present in the azurophil granules of polymorphonuclear leukocytes along with elastase and cathepsin G. Proteinase-3 gene expression is confined to the promyelocytic stage of polymorphonuclear leukocyte maturation. The present investigation identifies elements responsible for this highly controlled tissue- and developmental-specific expression of proteinase-3. Within the first 200 base pairs of the proteinase-3 promoter, two elements were identified as important for expression, these elements at -101 and -190 confer the majority of the activity. The element at -101 has a PU.1 consensus. It binds a myeloid nuclear protein of approximately 45 kDa that "supershifts" with PU.1 antibody and is competed by the CD11b PU.1 element. The element at -190 has a core sequence of CCCCGCCC (CG element). The cytidines but not the guanidine are essential for promoter activity. The CG element binds a second nuclear protein with a molecular mass of approximately 40 kDa that is found in cells of myeloid lineage as well as non-myeloid HeLa cells. However, the proteinase-3 promoter is not active in HeLa cells which suggests that the CG element alone is not sufficient for proteinase-3 gene expression. Maturation of promyelocytic cells results in an inhibition of proteinase-3 gene expression and a reduction in nuclear protein binding to the PU.1 and CG elements. Similar elements occur in the elastase and cathepsin G promoters. Using the elastase and cathepsin G PU.1 and CG-like elements as probes results in identical band-shift patterns to that obtained with proteinase-3 PU.1 and CG elements. These data suggest that there is cooperative interaction between a PU.1 and a CG element with a consensus of CCCCXCCC and that they are important control elements for tissue- and developmental-specific expression of azurophil serine proteinases of polymorphonuclear leukocytes.
Collapse
Affiliation(s)
- A Sturrock
- Department of Internal Medicine, Division of Respiratory, Critical Care, and Occupational Medicine, University of Utah Health Science Center, Salt Lake City, Utah 84132, USA
| | | | | |
Collapse
|
23
|
Abstract
The responses of leukemic cells to recombinant hematopoietic growth factors (HGFs) have been extensively studied, both in vitro and in vivo. Here, we summarize the current knowledge of the role that HGFs and growth modulatory cytokines play in the growth of leukemia. Particular attention is paid to the proliferation and maturation abilities of acute myeloblastic leukemia (AML), and the role HGFs play in these processes. The HGF responses of leukemic cells are discussed in the context of autocrine growth mechanisms, (cyto-)genetic abnormalities and defective function of HGF receptors. These studies have contributed considerably to our insight into the heterogeneous pathophysiology of leukemia.
Collapse
Affiliation(s)
- I P Touw
- Department of Hematology, Dr. Daniel den Hoed Cancer Center, Erasmus University, Rotterdam, The Netherlands
| | | |
Collapse
|
24
|
He Y, Borellini F, Koch WH, Huang KX, Glazer RI. Transcriptional regulation of c-Fes in myeloid leukemia cells. BIOCHIMICA ET BIOPHYSICA ACTA 1996; 1306:179-86. [PMID: 8634335 DOI: 10.1016/0167-4781(96)00005-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The c-Fes proto-oncogene encodes a myeloid-specific protein-tyrosine kinase that is expressed preferentially in differentiated myeloid cells, but not in early myeloblast progenitor cells. To examine the basis for the phenotypic expression of c-Fes, the transcription initiation sites of the human c-Fes gene were mapped in myeloid leukemia cells and regulatory elements in the genomic c-Fes sequence were characterized. Two major transcription initiation sites were found in the myeloid leukemia cell line THP-1 which delineated exon 1 to be 72-83 bp. When the activity of the CAT reporter gene under the control of the c-Fes promoter region, untranslated exon 1 and intron 1 was measured in TF-1, K562 and MCF-7 cells, only TF-1 cells exhibited chloramphenicol acetyltransferase activity. In contrast, all cell lines supported reporter gene activity when intron 1 was deleted. Deletion analyses revealed a negative regulatory region in intron 1, which was localized by Southwestern analysis and DNA footprinting to a 14 bp region. This negative regulatory region suppressed reporter CAT activity in K562 and TF-1 cells when inserted downstream to the SV40 early promoter. These results suggest that the tissue-specific expression of c-Fes may result, in part, from the negative regulation of transcription in myeloid and nonmyeloid cells.
Collapse
Affiliation(s)
- Y He
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC 20007, USA
| | | | | | | | | |
Collapse
|
25
|
Abstract
The study of oncogenes has provided numerous insights, not only into the mechanisms by which growth regulation becomes uncontrolled in cancer cells, but also into signal transduction processes which regulate the orderly proliferation and maturation of cells. c-fes/fps is a cellular oncogene which has been transduced frequently by mammalian and avian retroviruses. There are several features about Fes which suggest it may play a unique role in myeloid cell growth and differentiation. While it contains a tyrosine kinase and SH2 domain, there is no SH3 domain or carboxy terminal regulatory phosphotyrosine such as found in the Src family of kinases. Fes has a unique N-terminal domain of over 400 amino acids of unknown function. It has been implicated in signaling by a variety of hematopoietic growth factors, and is predominantly a nuclear protein.
Collapse
Affiliation(s)
- K E Yates
- Department of Medicine, UCLA School of Medicine 90095-1678, USA
| | | |
Collapse
|
26
|
Davies M, Harris S, Rudland P, Barraclough R. Expression of the rat, S-100-related, calcium-binding protein gene, p9Ka, in transgenic mice demonstrates different patterns of expression between these two species. DNA Cell Biol 1995; 14:825-32. [PMID: 7546288 DOI: 10.1089/dna.1995.14.825] [Citation(s) in RCA: 28] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
p9Ka (also known as mts1/18A2/calvasculin/CAPL) is a member of the S-100-related family of small, calcium-binding proteins. Previous studies suggest apparent discrepancies between the expression of the p9Ka gene in rat, mouse, and human tissues. Here we demonstrate that the natural p9Ka gene is expressed at lower levels in mouse than in rat, and that, in mouse but not in rat, p9Ka mRNA is more highly expressed in cells of lymphoid origin. Transgenic mouse strains express rat-p9Ka transgenes in a gene copy-number-dependent manner. The rat p9Ka transgene mRNA shows the same tissue distribution in several lines of transgenic mice, a distribution that is characteristic of the rat, from which the transgenes were derived. These results show that there is a difference in the pattern of expression of the same gene in two closely related species, and that the pattern of expression found in rat is specified by the DNA in the rat gene itself.
Collapse
Affiliation(s)
- M Davies
- Department of Biochemistry, University of Liverpool, UK
| | | | | | | |
Collapse
|
27
|
Krnacik MJ, Li S, Liao J, Rosen JM. Position-independent expression of whey acidic protein transgenes. J Biol Chem 1995; 270:11119-29. [PMID: 7744742 DOI: 10.1074/jbc.270.19.11119] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The expression of a 3-kilobase genomic rat whey acidic protein (WAP) clone (-949/+2020) in transgenic mice has been demonstrated previously to be copy number-dependent and independent of the site of integration (Dale, T., Krnacik, M. J., Schmidhauser, C., Yang, C. Q.-L., Bissell, M. J., and Rosen, J. M. (1992) Mol. Cell. Biol. 12, 905-914). The present study demonstrated that position-independent expression of the rat WAP -949/+2020 transgene was dependent on transgene spacing. Position-independent expression also was inhibited by an internal replacement of 49 base pair within the conserved GC-rich 3'-untranslated region (3'-UTR) with an identically sized nonspecific DNA sequence. Using electrophoretic mobility shift assays, nuclear factors isolated from mouse and human cells were shown to associate specifically with the rWAP 3'-UTR DNA, but not with the 3'-UTR containing the internal replacement or specific point mutations. Since a single copy of the 3'-UTR inserted 5' of the promoter could not rescue the 3'-UTR deletion, the 3'-UTR element does not appear to be functioning as either a classic enhancer or insulator element. However, the level of expression of rWAP transgenes was correlated with transgene association with the chromosomal scaffold in vivo.
Collapse
Affiliation(s)
- M J Krnacik
- Department of Cell Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
28
|
Abstract
In this review we discuss the concept of anti-angiogenesis, which is the inhibition of neovascularization. Anti-angiogenic agents are viewed from the standpoint of their effect on various elements of the angiogenic process, including induction of vascular discontinuity, endothelial cell movement, endothelial cell proliferation, and three-dimensional restructuring of patent vessels. An effort is made to place the many different approaches to anti-angiogenesis research into a comprehensible structure, in order to identify problems of evaluation and interpretation, thereby providing a clearer basis for determining promising and needed directions for further investigation.
Collapse
Affiliation(s)
- W Auerbach
- Center for Developmental Biology, University of Wisconsin, Madison 53706
| | | |
Collapse
|
29
|
Porter SD, Meyer CJ. A distal tyrosinase upstream element stimulates gene expression in neural-crest-derived melanocytes of transgenic mice: position-independent and mosaic expression. Development 1994; 120:2103-11. [PMID: 7925014 DOI: 10.1242/dev.120.8.2103] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have assessed the importance of a melanocyte-specific DNase I hypersensitive site and matrix attachment region situated 15 kb upstream of the mouse tyrosinase gene by analysis in transgenic mice. Transgenes containing all, part, or none of this region linked to the tyrosinase promoter and human tyrosinase cDNA were introduced into genetically albino mice, and pigmentation and transgene message levels were analyzed in the resulting transgenic lines. The effect of the upstream region was to enhance significantly gene expression in melanocytes, and to provide position-independent expression of the transgene. Two exceptions to complete position independence were seen; these lines displayed a mosaic expression pattern in which the transgene was expressed fully in some melanocyte clones but less so in others, resulting in transverse stripes of colours ranging from near white to dark grey. Unexpectedly, pigmentation in the eye of all transgenic lines containing the upstream region was nonuniform, in that the neural-crest-derived melanocytes of the choroid and anterior iris contained significantly more pigment than those derived from the optic cup (retinal pigment epithelium and posterior iris). Transgenes containing a small part or none of the upstream region were expressed poorly and in a position-dependent manner; of those lines that were visibly pigmented, expression was equal in the neural crest and optic-cup-derived cells of the eye.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- S D Porter
- Department of Pathology, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
30
|
Izuhara K, Feldman R, Greer P, Harada N. Interaction of the c-fes proto-oncogene product with the interleukin-4 receptor. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(17)32355-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
31
|
Abstract
In little more than a decade, the techniques developed for altering the genetic makeup of laboratory and livestock animals and plants have changed the landscape of biological research. It is now possible to introduce virtually any cloned gene into the germ line and study the expression pattern and effects of the introduced gene, or transgene. This has allowed the extension of in vitro and in vivo cell-culture studies into whole animal systems in which the introduced gene is subject to all normal regulatory processes from the onset of development. Although there have been reports of foreign gene expression resulting from direct injection of DNA in animals (e.g., Wolff et al., 1990; Zhu et al., 1993), transgenic animals are the primary model system for examining molecular genetic phenomena in vivo.
Collapse
Affiliation(s)
- D C Wight
- Edison Biotechnology Institute, Ohio University, Athens 45701
| | | |
Collapse
|
32
|
Arai Y, Kajihara S, Masuda J, Ohishi S, Zen K, Ogata J, Mukai T. Position-independent, high-level, and correct regional expression of the rat aldolase C gene in the central nervous system of transgenic mice. EUROPEAN JOURNAL OF BIOCHEMISTRY 1994; 221:253-60. [PMID: 8168514 DOI: 10.1111/j.1432-1033.1994.tb18736.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Aldolase C is mainly expressed in the central nervous system (CNS). To clarify the regulatory mechanisms for the CNS-specific expression, transgenic mice were created using two constructs of the rat aldolase C gene. A fusion gene comprising the 5' regulatory region of the aldolase C gene was expressed in a CNS-specific manner. However, the expression levels and the cellular localization of the gene varied among transgenic mice. The other construct, including both 5' and 3' regulatory regions of the gene, showed position-independent and high-level expression as well as the correct regional distribution in the CNS. These results indicate that the 13-kb sequence of the rat aldolase C gene contains sufficient information for faithful expression of the gene.
Collapse
Affiliation(s)
- Y Arai
- National Cardiovascular Center Research Institute, Suita, Japan
| | | | | | | | | | | | | |
Collapse
|
33
|
|
34
|
Quelle FW, Egerton M, Samelson LE, Wojchowski DM. Cytokine-induced phosphorylation of pp100 in FDC-ER cells is at tyrosine residues. Biochem Biophys Res Commun 1992; 188:1040-6. [PMID: 1280128 DOI: 10.1016/0006-291x(92)91336-o] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Using FDC-P1 cells stably transfected with a murine erythropoietin receptor cDNA as a model, we recently have shown that erythropoietin (EPO), IL-3 and GM-CSF each induce the rapid phosphorylation of a common cytosolic target, i.e., a M(r) 100,000 phosphoprotein "pp100". Presently, we demonstrate that cytokine-induced phosphorylation of pp100 is primarily at tyrosine residues. This is shown by Western blotting with the anti-phosphotyrosine antibody PY20, and by the resistance of [32P]-pp100 to hydroxide-mediated hydrolysis of phosphates. These data, together with the recent observation by Linnekin et al. that pp100/p97 apparently associates directly with EPO receptors, suggest that pp100 may comprise an immediate common component in the signal transduction pathways of EPO, IL-3, GM-CSF and possibly other type I/II cytokine receptors. Additional analyses suggest that pp100 is distinct from a previously described M(r) 100,000 cytosolic target which is tyrosine phosphorylated in hematopoietic cells upon activation of T-cell receptors.
Collapse
Affiliation(s)
- F W Quelle
- Department of Molecular and Cell Biology, Pennsylvania State University, University Park 16802
| | | | | | | |
Collapse
|
35
|
Ferrari S, Manfredini R, Grande A, Torelli U. Antisense strategies to characterize the role of genes and oncogenes involved in myeloid differentiation. Ann N Y Acad Sci 1992; 660:11-26. [PMID: 1340115 DOI: 10.1111/j.1749-6632.1992.tb21053.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- S Ferrari
- Experimental Hematology Center, II Medical Clinic, University of Modena, Italy
| | | | | | | |
Collapse
|
36
|
van Bokhoven A, van Duijnhoven HL, Jücker M, Roebroek AJ, van de Ven WJ. Development and characterization of a panel of monoclonal antibodies against the catalytic domain of the human fes proto-oncogene product. Mol Biol Rep 1992; 16:17-25. [PMID: 1545781 DOI: 10.1007/bf00788749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In developing monoclonal antibodies (Moabs) against the human fes proto-oncogene product, recombinant DNA technology was used to target reactivity of the Moabs towards the catalytic domain of it. Therefore, sequences of human fes exons 15-19 encoding amino acid residues 612 to 822 which harbor the catalytic domain except the presumed ATP-binding region, were fused in phase to the bacterial trp E gene which encodes anthranilate synthase. After partial purification of it, the bacterially produced hybrid product of this trp E-delta fes fusion gene was used as immunogen. A series of twelve mouse Moabs was obtained which recognized the human p92fes protein and the viral oncogene product p85gag-fes encoded by the Snyder-Theilen strain of feline sarcoma virus. Reactivity appeared to be directed towards the catalytic domain of the human fes proto-oncogene product. This was demonstrated by in vitro transcription and translation experiments using human fes coding sequences from exons 16-19. Upon testing their functional activity in divers immunological techniques, the whole panel of Moabs appeared to be useful in immunoprecipitation, Western blot and immunohistochemical analysis. Immunocytochemical analysis indicated that p85gag-fes is predominantly a cytoplasmic protein.
Collapse
Affiliation(s)
- A van Bokhoven
- Department of Biochemistry, University of Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
37
|
Skalnik D, Strauss E, Orkin S. CCAAT displacement protein as a repressor of the myelomonocytic-specific gp91-phox gene promoter. J Biol Chem 1991. [DOI: 10.1016/s0021-9258(18)55363-1] [Citation(s) in RCA: 130] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
38
|
Shapiro L, Ashmun R, Roberts W, Look A. Separate promoters control transcription of the human aminopeptidase N gene in myeloid and intestinal epithelial cells. J Biol Chem 1991. [DOI: 10.1016/s0021-9258(18)99056-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
39
|
Adams JM, Cory S. Transgenic models for haemopoietic malignancies. BIOCHIMICA ET BIOPHYSICA ACTA 1991; 1072:9-31. [PMID: 2018781 DOI: 10.1016/0304-419x(91)90004-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- J M Adams
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | | |
Collapse
|