1
|
Simonini S, Bencivenga S, Grossniklaus U. A paternal signal induces endosperm proliferation upon fertilization in Arabidopsis. Science 2024; 383:646-653. [PMID: 38330116 DOI: 10.1126/science.adj4996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 01/04/2024] [Indexed: 02/10/2024]
Abstract
In multicellular organisms, sexual reproduction relies on the formation of highly differentiated cells, the gametes, which await fertilization in a quiescent state. Upon fertilization, the cell cycle resumes. Successful development requires that male and female gametes are in the same phase of the cell cycle. The molecular mechanisms that reinstate cell division in a fertilization-dependent manner are poorly understood in both animals and plants. Using Arabidopsis, we show that a sperm-derived signal induces the proliferation of a female gamete, the central cell, precisely upon fertilization. The central cell is arrested in S phase by the activity of the RETINOBLASTOMA RELATED1 (RBR1) protein. Upon fertilization, delivery of the core cell cycle component CYCD7;1 causes RBR1 degradation and thus S phase progression, ensuring the formation of functional endosperm and, consequently, viable seeds.
Collapse
Affiliation(s)
- Sara Simonini
- Institute of Plant and Microbial Biology and Zurich-Basel Plant Science Center, University of Zurich, CH-8008 Zurich, Switzerland
| | - Stefano Bencivenga
- Institute of Plant and Microbial Biology and Zurich-Basel Plant Science Center, University of Zurich, CH-8008 Zurich, Switzerland
| | - Ueli Grossniklaus
- Institute of Plant and Microbial Biology and Zurich-Basel Plant Science Center, University of Zurich, CH-8008 Zurich, Switzerland
| |
Collapse
|
2
|
Mai S, Hodges A, Chen HM, Zhang J, Wang YL, Liu Y, Nakatsu F, Wang X, Fang J, Xu Y, Davidov V, Kang K, Pingali SR, Ganguly S, Suzuki M, Konopleva M, Prinzing B, Zu Y, Gottschalk S, Lu Y, Chen SH, Pan PY. LILRB3 Modulates Acute Myeloid Leukemia Progression and Acts as an Effective Target for CAR T-cell Therapy. Cancer Res 2023; 83:4047-4062. [PMID: 38098451 DOI: 10.1158/0008-5472.can-22-2483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 04/11/2023] [Accepted: 06/14/2023] [Indexed: 12/18/2023]
Abstract
Identifying novel cell surface receptors that regulate leukemia cell differentiation and can be targeted to inhibit cellular proliferation is crucial to improve current treatment modalities in acute myeloid leukemia (AML), especially for relapsed or chemotherapy-refractory leukemia. Leukocyte immunoglobulin-like receptor type B (LILRB) is an immunomodulatory receptor originally found to be expressed in myeloid cells. In this study, we found that LILRB receptors can be induced under inflammatory stimuli and chemotherapy treatment conditions. Blockade of LILRB3 inhibited leukemia cell proliferation and leukemia progression. In addition, treatment with LILRB3 blocking antibodies upregulated myeloid lineage differentiation transcription factors, including PU.1, C/EBP family, and IRF, whereas phosphorylation of proliferation regulators, for example, AKT, cyclin D1, and retinoblastoma protein, was decreased. Conversely, transcriptomic analysis showed LILRB3 activation by agonist antibodies may enhance leukemia survival through upregulation of cholesterol metabolism, which has been shown to promote leukemia cell survival. Moreover, LILRB3-targeted CAR T cells exhibited potent antitumor effects both in vitro and in vivo. Taken together, our results suggest that LILRB3 is a potentially potent target for multiple treatment modalities in AML. SIGNIFICANCE LILRB3 regulates differentiation and proliferation in acute myeloid leukemia and can be targeted with monoclonal antibodies and CAR T cells to suppress leukemia growth.
Collapse
Affiliation(s)
- Sunny Mai
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas
| | - Alan Hodges
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas
- Texas A&M University System School of Medicine, Bryan, Texas
| | - Hui-Ming Chen
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas
| | - Jilu Zhang
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas
| | - Yi-Ling Wang
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas
| | - Yongbin Liu
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas
| | - Fumiko Nakatsu
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Xiaoxuan Wang
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas
| | - Jing Fang
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas
| | - Yitian Xu
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas
| | - Vitaliy Davidov
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas
- Texas A&M University System School of Medicine, Bryan, Texas
| | - Kyeongah Kang
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas
| | - Sai Ravi Pingali
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas
- Division of Hematology, Medical Oncology and Hematology, Houston Methodist Hospital, Houston, Texas
| | - Siddhartha Ganguly
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas
- Division of Hematology, Medical Oncology and Hematology, Houston Methodist Hospital, Houston, Texas
| | - Masataka Suzuki
- Center for Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Marina Konopleva
- Department of Oncology, Albert Einstein College of Medicine, Bronx, New York
| | - Brooke Prinzing
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Youli Zu
- Department of Pathology & Genomic Medicine, Houston Methodist Research Institute, Houston Texas
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation & Cellular Therapy, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Yong Lu
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas
| | - Shu-Hsia Chen
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas
- Texas A&M University System School of Medicine, Bryan, Texas
- Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medical Science and Graduate School of Medical Sciences, New York, New York
| | - Ping-Ying Pan
- Center for Immunotherapy, Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas
- Texas A&M University System School of Medicine, Bryan, Texas
| |
Collapse
|
3
|
Rhus coriaria induces senescence and autophagic cell death in breast cancer cells through a mechanism involving p38 and ERK1/2 activation. Sci Rep 2015; 5:13013. [PMID: 26263881 PMCID: PMC4532997 DOI: 10.1038/srep13013] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/13/2015] [Indexed: 12/29/2022] Open
Abstract
Here, we investigated the anticancer effect of Rhus coriaria on three breast cancer cell lines. We demonstrated that Rhus coriaria ethanolic extract (RCE) inhibits the proliferation of these cell lines in a time- and concentration-dependent manner. RCE induced senescence and cell cycle arrest at G1 phase. These changes were concomitant with upregulation of p21, downregulation of cyclin D1, p27, PCNA, c-myc, phospho-RB and expression of senescence-associated β-galactosidase activity. No proliferative recovery was detected after RCE removal. Annexin V staining and PARP cleavage analysis revealed a minimal induction of apoptosis in MDA-MB-231 cells. Electron microscopy revealed the presence of autophagic vacuoles in RCE-treated cells. Interestingly, blocking autophagy by 3-methyladenine (3-MA) or chloroquine (CQ) reduced RCE-induced cell death and senescence. RCE was also found to activate p38 and ERK1/2 signaling pathways which coincided with induction of autophagy. Furthermore, we found that while both autophagy inhibitors abolished p38 phosphorylation, only CQ led to significant decrease in pERK1/2. Finally, RCE induced DNA damage and reduced mutant p53, two events that preceded autophagy. Our findings provide strong evidence that R. coriaria possesses strong anti-breast cancer activity through induction of senescence and autophagic cell death, making it a promising alternative or adjunct therapeutic candidate against breast cancer.
Collapse
|
4
|
Fulcher AJ, Dias MM, Jans DA. Binding of p110 retinoblastoma protein inhibits nuclear import of simian virus SV40 large tumor antigen. J Biol Chem 2010; 285:17744-53. [PMID: 20356831 PMCID: PMC2878538 DOI: 10.1074/jbc.m109.055491] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2009] [Revised: 03/29/2010] [Indexed: 01/19/2023] Open
Abstract
Nuclear import of the simian virus 40 large tumor antigen (T-ag) is dependent on its nuclear localization signal (NLS) within amino acids 126-132 that is recognized by the importin alpha/beta1 heterodimer, as well as a protein kinase CK2 site at serine 112 upstream of the NLS, which enhances the interaction approximately 50-fold. Here we show for the first time that T-ag nuclear import is negatively regulated by N-terminal sequences (amino acids 102-110), which represent the binding site (BS) for the retinoblastoma (Rb) tumor suppressor protein (p110(Rb)). Quantitative confocal laser scanning microscopic analysis of the transport properties of T-ag constructs with or without Rb binding site mutations in living transfected cells or in a reconstituted nuclear transport system indicates that the presence of the RbBS significantly reduces nuclear accumulation of T-ag. A number of approaches, including the analysis of T-ag nuclear import in an isogenic cell pair with and without functional p110(Rb) implicate p110(Rb) binding as being responsible for the reduced nuclear accumulation, with the Ser(106) phosphorylation site within the RbBS appearing to enhance the inhibitory effect. Immunoprecipitation experiments confirmed association of T-ag and p110(Rb) and dependence thereof on negative charge at Ser(106). The involvement of p110(Rb) in modulating T-ag nuclear transport has implications for the regulation of nuclear import of other proteins from viruses of medical significance that interact with p110(Rb), and how this may relate to transformation.
Collapse
Affiliation(s)
- Alex James Fulcher
- From the Nuclear Signaling Laboratory, Department of Biochemistry and Molecular Biology, Monash University, Victoria, Clayton 3800, Australia and
| | - Manisha M. Dias
- From the Nuclear Signaling Laboratory, Department of Biochemistry and Molecular Biology, Monash University, Victoria, Clayton 3800, Australia and
| | - David A. Jans
- From the Nuclear Signaling Laboratory, Department of Biochemistry and Molecular Biology, Monash University, Victoria, Clayton 3800, Australia and
- the ARC Centre of Excellence for Biotechnology and Development, Victoria, Melbourne 3000, Australia
| |
Collapse
|
5
|
Luyckx VA, Compston CA, Simmen T, Mueller TF. Accelerated senescence in kidneys of low-birth-weight rats after catch-up growth. Am J Physiol Renal Physiol 2009; 297:F1697-705. [PMID: 19828676 DOI: 10.1152/ajprenal.00462.2009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Epidemiological studies show a strong association between low birth weight and hypertension, renal, and cardiovascular disease, especially after catch-up growth. Senescence is an important contributor to the progression of chronic disease. Developmentally programmed premature senescence may be a link among low birth weight, catch-up growth, and adult disease. Low birth weight was induced by feeding pregnant rats a low-protein diet from day 12 of gestation to 10 days postdelivery. Low- and normal-birth-weight male offspring were weaned onto regular or high-calorie diets to enhance catch-up growth. Kidneys and hearts of offspring were analyzed for RNA and protein markers of stress-induced senescence (p16, p21, p53, Rb). Markers of mitochondrial stress (p66Shc) and activation of endoplasmic reticulum protein secretion (Ero1alpha) were analyzed as regulators of reactive oxygen species generation. Reactive oxygen species are known to be associated with premature aging. Senescence markers were not different in low- or normal-birth-weight kidneys at birth. During rapid catch-up growth, p16 and p21 increased significantly in low-birth-weight kidneys and hearts (P < 0.01). Renal p16 levels increased progressively and were significantly higher in low-birth-weight kidneys at 3 and 6 mo (P < or = 0.02). Renal p66Shc and Ero1alpha were significantly higher in low- compared with normal- birth-weight kidneys at 6 mo, suggesting reactive oxygen species generation (P < or = 0.03). Low-birth-weight rats exhibit accelerated senescence in kidneys and hearts after rapid catch-up growth, a likely important link between early growth and subsequent hypertension, renal, and cardiovascular disease.
Collapse
Affiliation(s)
- Valerie A Luyckx
- Division of Nephrology, University of Alberta, Edmonton, Alberta, Canada.
| | | | | | | |
Collapse
|
6
|
Dasgupta B, Milbrandt J. AMP-activated protein kinase phosphorylates retinoblastoma protein to control mammalian brain development. Dev Cell 2009; 16:256-70. [PMID: 19217427 DOI: 10.1016/j.devcel.2009.01.005] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Revised: 10/29/2008] [Accepted: 01/20/2009] [Indexed: 12/22/2022]
Abstract
AMP-activated protein kinase (AMPK) is an evolutionarily conserved metabolic sensor that responds to alterations in cellular energy levels to maintain energy balance. While its role in metabolic homeostasis is well documented, its role in mammalian development is less clear. Here we demonstrate that mutant mice lacking the regulatory AMPK beta1 subunit have profound brain abnormalities. The beta1(-/-) mice show atrophy of the dentate gyrus and cerebellum, and severe loss of neurons, oligodendrocytes, and myelination throughout the central nervous system. These abnormalities stem from reduced AMPK activity, with ensuing cell cycle defects in neural stem and progenitor cells (NPCs). The beta1(-/-) NPC deficits result from hypophosphorylation of the retinoblastoma protein (Rb), which is directly phosphorylated by AMPK at Ser(804). The AMPK-Rb axis is utilized by both growth factors and energy restriction to increase NPC growth. Our results reveal that AMPK integrates growth factor signaling with cell cycle control to regulate brain development.
Collapse
Affiliation(s)
- Biplab Dasgupta
- Department of Pathology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | |
Collapse
|
7
|
Balint BL, Szanto A, Madi A, Bauer UM, Gabor P, Benko S, Puskás LG, Davies PJA, Nagy L. Arginine methylation provides epigenetic transcription memory for retinoid-induced differentiation in myeloid cells. Mol Cell Biol 2005; 25:5648-63. [PMID: 15964820 PMCID: PMC1156990 DOI: 10.1128/mcb.25.13.5648-5663.2005] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cellular differentiation is governed by changes in gene expression, but at the same time, a cell's identity needs to be maintained through multiple cell divisions during maturation. In myeloid cell lines, retinoids induce gene expression and a well-characterized two-step lineage-specific differentiation. To identify mechanisms that contribute to cellular transcriptional memory, we analyzed the epigenetic changes taking place on regulatory regions of tissue transglutaminase, a gene whose expression is tightly linked to retinoid-induced differentiation. Here we report that the induction of an intermediary or "primed" state of myeloid differentiation is associated with increased H4 arginine 3 and decreased H3 lysine 4 methylation. These modifications occur before transcription and appear to prime the chromatin for subsequent hormone-regulated transcription. Moreover, inhibition of methyltransferase activity, pre-acetylation, or activation of the enzyme PAD4 attenuated retinoid-regulated gene expression, while overexpression of PRMT1, a methyltransferase, enhanced retinoid responsiveness. Taken together, our results suggest that H4 arginine 3 methylation is a bona fide positive epigenetic marker and regulator of transcriptional responsiveness as well as a signal integration mechanism during cell differentiation and, as such, may provide epigenetic memory.
Collapse
Affiliation(s)
- Balint L Balint
- Department of Biochemistry and Molecular Biology, Research Center for Molecular Medicine, University of Debrecen, Medical and Health Science Center, Nagyerdei krt. 98, Debrecen H-4012, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Flaminio MJBF, Yen A, Antczak DF. The proliferation inhibitory proteins p27Kip1 and retinoblastoma are involved in the control of equine lymphocyte proliferation. Vet Immunol Immunopathol 2004; 102:363-77. [PMID: 15541790 DOI: 10.1016/j.vetimm.2004.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2004] [Revised: 06/30/2004] [Accepted: 07/22/2004] [Indexed: 11/19/2022]
Abstract
Observations in early equine pregnancy clearly reveal maternal immune recognition of and response to the presence of the conceptus. Nevertheless, both maternal cellular and humoral responses appear ineffective in destroying the developing placenta and fetus in early pregnancy. Our previous studies had shown that the pre-conditioned medium generated from the culture of equine invasive trophoblast inhibited mitogen-induced lymphocyte proliferation and the expression of cytokine messenger RNA in vitro. Those findings also suggested that lymphocytes might have been halted in the G0/G1 phase of the cell cycle. To characterize the cell cycle and the intracellular mechanisms involved in the inhibition of lymphocyte proliferation, equine peripheral blood lymphocytes were cultured in the presence or absence of pokeweed mitogen (PWM) in fresh medium, or in medium pre-conditioned through cell culture of invasive trophoblast cells or fetal fibroblasts. Two-color flow cytometric analysis for bromodeoxyuridine (BrdU) incorporation by stimulated lymphocytes, and concomitant DNA staining with 7-amino-actinomycin D (7-AAD), indicated that a greater proportion of lymphocytes were found in the G0/G1 phase of the cell cycle when cultured in the invasive trophoblast cell pre-conditioned medium compared to controls. Analysis using carboxyfluorescein diacetate succinimidyl ester (CFSE) fluorescence intensity demonstrated that lymphocytes cultured in the presence of invasive trophoblast cell pre-conditioned medium had fewer cells going through division, but that those fewer cells sustained similar numbers of cell divisions as in control cultures. Hypophosphorylated retinoblastoma (Rb) protein expression was increased and p27Kip1 expression was maintained at higher levels in lymphocytes cultured in invasive trophoblast pre-conditioned medium compared to fresh medium. In agreement with these data, flow cytometric measurement of the Ki-67 protein expression in lymphocytes cultured in invasive trophoblast pre-conditioned medium was lower in comparison to controls. These findings suggest that the equine lymphocyte proliferation is at least partially regulated by the expression of proliferation inhibitory proteins such as p27Kip1 and hypophosphorylated Rb. These proteins seem to be important regulators of cell cycle transition between G1 and S phase in equine lymphocytes.
Collapse
Affiliation(s)
- M Julia B F Flaminio
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| | | | | |
Collapse
|
9
|
Roig JM, Molina MA, Cascante A, Calbó J, Carbó N, Wirtz U, Sreedharan S, Fillat C, Mazo A. Adenovirus-mediated retinoblastoma 94 gene transfer induces human pancreatic tumor regression in a mouse xenograft model. Clin Cancer Res 2004; 10:1454-62. [PMID: 14977849 DOI: 10.1158/1078-0432.ccr-0442-03] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Gene transfer of a truncated variant of the retinoblastoma (RB) gene encoding a M(r) 94000 protein that lacks the NH(2)-terminal 112 amino acid residues, termed RB94, has been shown to inhibit proliferation of several human tumor cell types. We have assessed its therapeutic effectiveness on pancreatic cancer, one of the most aggressive and therapy-resistant types of cancer. For this purpose, preclinical studies aimed to evaluate the therapeutic potential of RB94 gene transfer in pancreatic cancer were carried out. EXPERIMENTAL DESIGN We have compared the antiproliferative effects of adenovirus-mediated gene transfer of RBwt and RB94 at the in vitro and in vivo levels in three RB-positive human pancreatic tumor cell lines: (a). NP-9; (b). NP-18; and (c). NP-31. We have also examined their effects on cell cycle and their capacity to induce apoptosis. RESULTS In vitro results indicate that RB94 gene transfer has stronger antiproliferative effects compared with RBwt. RB94 transduction correlated with accumulation at the S-G(2) phase of the cell cycle in the three cell lines tested and induction of apoptosis in two of them. In vivo studies show significant decreases in the growth rate of tumors treated with Ad-RB94 when compared with those treated with Ad-RBwt. Moreover, terminal deoxynucleotidyl transferase-mediated nick end labeling analyses of Ad-RB94-treated tumor sections revealed that only RB94 is able to significantly induce apoptosis. CONCLUSIONS RB94 gene expression has antiproliferative effects also in human pancreatic tumor cells, being more effective than wild-type RB in preventing tumor growth.
Collapse
Affiliation(s)
- Josep Maria Roig
- Department of Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Rorie CJ, Weissman BE. The Ews/Fli-1 Fusion Gene Changes the Status of p53 in Neuroblastoma Tumor Cell Lines. Cancer Res 2004; 64:7288-95. [PMID: 15492248 DOI: 10.1158/0008-5472.can-04-1610] [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] [Indexed: 11/16/2022]
Abstract
One hallmark of Ewing's sarcoma/peripheral neuroectodermal tumors is the presence of the Ews/Fli-1 chimeric oncogene. Interestingly, infection of neuroblastoma tumor cell lines with Ews/Fli-1 switches the differentiation program of neuroblastomas to Ewing's sarcoma/peripheral neuroectodermal tumors. Here we examined the status of cytoplasmically sequestered wt-p53 in neuroblastomas after stable expression of Ews/Fli-1. Immunofluorescence revealed that in the neuroblastoma-Ews/Fli-1 infectant cell lines, p53 went from a punctate-pattern of cytoplasmic sequestration to increased nuclear localization. Western blot analysis revealed that PARC was down-regulated in one neuroblastoma cell line but not expressed in the second. Therefore, decreased PARC expression could not fully account for relieving p53 sequestration in the neuroblastoma tumor cells. Neuroblastoma-Ews/Fli-1 infectant cell lines showed marked increases in p53 protein expression without transcriptional up-regulation. Interestingly, p53 was primarily phosphorylated, without activation of its downstream target p21(WAF1). Western blot analysis revealed that whereas MDM2 gene expression does not change, p14(ARF), a negative protein regulator of MDM2, increases. These observations suggest that the downstream p53 pathway may be inactivated as a result of abnormal p53. We also found that p53 has an extended half-life in the neuroblastoma-Ews/Fli-1 infectants despite the retention of a wild-type sequence in neuroblastoma-Ews/Fli-1 infectant cell lines. We then tested the p53 response pathway and observed that the neuroblastoma parent cells responded to genotoxic stress, whereas the neuroblastoma-Ews/Fli-1 infectants did not. These results suggest that Ews/Fli-1 can directly abrogate the p53 pathway to promote tumorigenesis. These studies also provide additional insight into the relationship among the p53 pathway proteins.
Collapse
Affiliation(s)
- Checo J Rorie
- Curriculum in Toxicology and Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | |
Collapse
|
11
|
Abstract
Retinoblastoma gene (Rb) is the prototype of tumor suppressors. Germline mutation in the retinoblastoma gene is susceptible to cancer and reintroduction of wild-type Rb is able to suppress neoplastic phenotypes. The fundamental cellular functions of Rb in the control of cell growth and differentiation are important for its tumor suppression. In general, cancer susceptibility caused by inactivation of a tumor suppressor gene results from genome instability. Accordingly, Rb may function in the maintenance of chromosome stability by influencing mitotic progression, faithful chromosome segregation, and structural remodeling of mitotic chromosomes. Rb is also implicated in the regulation of replication machinery and in the control of cell cycle checkpoints in response to DNA damage, further supporting such a role for Rb. Moreover, the mechanistic basis for Rb-mediated transcriptional repression has revealed its connection to global chromatin remodeling. It is likely that Rb suppresses tumor formation by virtue of its multiple biological activities, and a theme throughout its multiple cellular functions is its central role in controlling activities that involve chromatin remodeling. A model in which Rb controls global genome fluidity is thus proposed. Finally, a recent study provides direct evidence indicating that loss of Rb function leads to genome instability. Therefore, tumor suppressors have a common role in the maintenance of genome stability, and such a role may be pivotal for their functions in tumor suppression.
Collapse
Affiliation(s)
- Lei Zheng
- Department of Molecular Medicine/Institute of Biotechnology, University of Texas Health Science Center at San Antonio, 78245, USA
| | | |
Collapse
|
12
|
Gottifredi V, Karni-Schmidt O, Shieh SS, Prives C. p53 down-regulates CHK1 through p21 and the retinoblastoma protein. Mol Cell Biol 2001; 21:1066-76. [PMID: 11158294 PMCID: PMC99561 DOI: 10.1128/mcb.21.4.1066-1076.2001] [Citation(s) in RCA: 135] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Both fission yeast and mammalian cells require the function of the checkpoint kinase CHK1 for G2 arrest after DNA damage. The tumor suppressor p53, a well-studied stress response factor, has also been shown to play a role in DNA damage G2 arrest, although in a manner that is probably independent of CHK1. p53, however, can be phosphorylated and regulated by both CHK1 as well as another checkpoint kinase, hCds1 (also called CHK2). It was therefore of interest to determine whether reciprocally, p53 affects either CHK1 or CHK2. We found that induction of p53 either by diverse stress signals or ectopically using a tetracycline-regulated promoter causes a marked reduction in CHK1 protein levels. CHK1 downregulation by p53 occurs as a result of reduced CHK1 RNA accumulation, indicating that repression occurs at the level of transcription. Repression of CHK1 by p53 requires p21, since p21 alone is sufficient for this to occur and cells lacking p21 cannot downregulate CHK1. Interestingly, pRB is also required for CHK1 downregulation, suggesting the possible involvement of E2F-dependent transcription in the regulation of CHK1. Our results identify a new repression target of p53 and suggest that p53 and CHK1 play interdependent and complementary roles in regulating both the arrest and resumption of G2 after DNA damage.
Collapse
Affiliation(s)
- V Gottifredi
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | | | | | | |
Collapse
|
13
|
Flatt PM, Tang LJ, Scatena CD, Szak ST, Pietenpol JA. p53 regulation of G(2) checkpoint is retinoblastoma protein dependent. Mol Cell Biol 2000; 20:4210-23. [PMID: 10825186 PMCID: PMC85790 DOI: 10.1128/mcb.20.12.4210-4223.2000] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In the present study, we investigated the role of p53 in G(2) checkpoint function by determining the mechanism by which p53 prevents premature exit from G(2) arrest after genotoxic stress. Using three cell model systems, each isogenic, we showed that either ectopic or endogenous p53 sustained a G(2) arrest activated by ionizing radiation or adriamycin. The mechanism was p21 and retinoblastoma protein (pRB) dependent and involved an initial inhibition of cyclin B1-Cdc2 activity and a secondary decrease in cyclin B1 and Cdc2 levels. Abrogation of p21 or pRB function in cells containing wild-type p53 blocked the down-regulation of cyclin B1 and Cdc2 expression and led to an accelerated exit from G(2) after genotoxic stress. Thus, similar to what occurs in p21 and p53 deficiency, pRB loss can uncouple S phase and mitosis after genotoxic stress in tumor cells. These results indicate that similar molecular mechanisms are required for p53 regulation of G(1) and G(2) checkpoints.
Collapse
Affiliation(s)
- P M Flatt
- Department of Biochemistry, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | | | | | | |
Collapse
|
14
|
Yen A, Varvayanis S. Retinoic acid increases amount of phosphorylated RAF; ectopic expression of cFMS reveals that retinoic acid-induced differentiation is more strongly dependent on ERK2 signaling than induced GO arrest is. In Vitro Cell Dev Biol Anim 2000; 36:249-55. [PMID: 10852350 DOI: 10.1290/1071-2690(2000)036<0249:raiaop>2.0.co;2] [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: 11/11/2022]
Abstract
Retinoic acid is known to cause the myeloid differentiation and G1/0 cell cycle arrest of HL-60 cells in a process that requires mitogen-activated protein/extracellular signal regulated kinase (MEK)-dependent extracellular signal regulated kinase (ERK)2 activation. It has also been shown that ectopic expression of cFMS, a platelet-derived growth factor (PDGF)-family transmembrane tyrosine kinase receptor, enhances retinoic acid-induced differentiation and G1/0 arrest. The mechanism of how the retinoic acid and cFMS signaling pathways intersect is not known. The present data show that the ectopic expression of cFMS results in the differential loss of sensitivity of retinoic acid-induced differentiation or G1/0 arrest to inhibition of ERK2 activation. PD98059 was used to inhibit MEK and consequently ERK2. In wildtype HL-60 cells, PD98059 blocked retinoic acid-induced differentiation; but in cFMS stable transfectants, PD98059 only attenuated the induced differentiation, with the resulting response resembling that of retinoic acid-treated wild-type HL-60. In wild-type HL-60, PD98059 greatly attenuated the retinoic acid-induced G1/0 arrest allied with retinoblastoma (RB) hypophosphorylation; but in cFMS stable transfectants, PD98059 had no inhibitory effect on RB hypophosphorylation and G1/0 arrest. This differential sensitivity to PD98059 and uncoupling of retinoic acid-induced differentiation and G1/0 arrest in cFMS transfectants is associated with changes in mitogen-activated protein kinase signaling molecules. The cFMS transfectants had more activated ERK2 than did the wild-type cells, which surprisingly was not attributable to enhanced mitogen-activated protein-kinase-kinase-kinase (RAF) phosphorylation. Retinoic acid increased the amount of activated ERK2 and phosphorylated RAF in both cell lines. But PD98059 eliminated detectable ERK2 activation, as well as inhibited RAF phosphorylation, in untreated and retinoic acid-treated wild-type HL-60 and cFMS transfectants, consistent with MEK or ERK feedback-regulation of RAF, in all four cases. Since PD98059 blocks the cFMS-conferred enhancement of the retinoic acid-induced differentiation, but not growth arrest, the data indicate that cFMS-enhanced differentiation acts primarily through MEK and ERK2, but cFMS-enhanced G1/0 arrest allied with RB hypophosphorylation depends on another cFMS signal route, which by itself can effect G1/0 arrest without activated ERK2. Ectopic expression of cFMS and differential sensitivity to ERK2 inhibition thus reveal that retinoic acid-induced HL-60 cell differentiation and G1/0 arrest are differentially dependent on ERK2 and can be uncoupled. A significant unanticipated finding was that retinoic acid caused a MEK-dependent increase in the amount of phosphorylated RAF. This increase may help sustain prolonged ERK2 activation.
Collapse
Affiliation(s)
- A Yen
- Departmernt of Biomredical Sciences, College of Veterinary Medicine, Cornell University Ithaca, New York 14853, USA.
| | | |
Collapse
|
15
|
Rigberg DA, Kim FS, Sebastian JL, Kazanjian KK, McFadden DW. Hypophosphorylated retinoblastoma protein is associated with G2 arrest in esophageal squamous cell carcinoma. J Surg Res 1999; 84:101-5. [PMID: 10334897 DOI: 10.1006/jsre.1999.5617] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hypophosphorylated retinoblastoma (Rb) gene product binds critical transcription factors, leading to G1 arrest in a number of conditions, including following DNA damage. We have previously shown that irradiated esophageal squamous cell carcinoma (ESSC) cells undergo predominantly G2 arrest, with increases in inhibitors of Rb phosphorylation. We thus hypothesized that this G2 arrest would be accompanied by increases in hypophosphorylated Rb protein (pRb). We sequenced the Rb genes of three human ESSC lines (KYSE) following reverse transcription polymerase chain reaction of exons A-E. Western gels were performed on protein extracts for pRb. Cells were irradiated at 6 Gy, and protein was extracted at 6 h. ELISA was used to measure hypophosphorylated pRb in radiated versus control cells. Student's t test was used to compare results. All lines had wild-type Rb genes. Western gels confirmed the presence of pRb. There were significant increases in hypophosphorylated pRb in all three lines following irradiation (no line with less than a 100% increase). We have thus shown that irradiation-induced G2 arrest occurs in association with wild-type Rb genes and that there is associated hypophosphorylation of pRb. This supports our data describing a further role for other G1 mediators, such as p21, in G2 arrest. Further investigations into therapies to expoit this cell cycle checkpoint are warranted and planned.
Collapse
Affiliation(s)
- D A Rigberg
- Department of Surgery, UCLA School of Medicine, Los Angeles, California, 90095, USA
| | | | | | | | | |
Collapse
|
16
|
|