Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
6.
J Virol ; 89(21): 10821-31, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26292318

ABSTRACT

UNLABELLED: Gammaherpesviruses (GHVs) carry homologs of cellular genes, including those encoding a viral cyclin that promotes reactivation from latent infection. The viral cyclin has reduced sensitivity to host cyclin-dependent kinase inhibitors in vitro; however, the in vivo significance of this is unclear. Here, we tested the genetic requirement for the viral cyclin in mice that lack the host inhibitors p27(Kip1) and p18(INK4c), two cyclin-dependent kinase inhibitors known to be important in regulating B cell proliferation and differentiation. While the viral cyclin was essential for reactivation in wild-type mice, strikingly, it was dispensable for reactivation in mice lacking p27(Kip1) and p18(INK4c). Further analysis revealed that genetic ablation of only p18(INK4c) alleviated the requirement for the viral cyclin for reactivation from latency. p18(INK4c) regulated reactivation in a dose-dependent manner so that the viral cyclin was dispensable in p18(INK4c) heterozygous mice. Finally, treatment of wild-type cells with the cytokine BAFF, a known attenuator of p18(INK4c) function in B lymphocytes, was also able to bypass the requirement for the viral cyclin in reactivation. These data show that the gammaherpesvirus viral cyclin functions specifically to bypass the cyclin-dependent kinase inhibitor p18(INK4c), revealing an unanticipated specificity between a GHV cyclin and a single cyclin-dependent kinase inhibitor. IMPORTANCE: The gammaherpesviruses (GHVs) cause lifelong infection and can cause chronic inflammatory diseases and cancer, especially in immunosuppressed individuals. Many GHVs encode a conserved viral cyclin that is required for infection and disease. While a common property of the viral cyclins is that they resist inhibition by normal cellular mechanisms, it remains unclear how important it is that the GHVs resist this inhibition. We used a mouse GHV that either contained or lacked a viral cyclin to test whether the viral cyclin lost importance when these inhibitory pathways were removed. These studies revealed that the viral cyclin was required for optimal function in normal mice but that it was no longer required following removal or reduced function of a single cellular inhibitor. These data define a very specific role for the viral cyclin in bypassing one cellular inhibitor and point to new methods to intervene with viral cyclins.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p18/metabolism , Cyclins/metabolism , Gammaherpesvirinae/metabolism , Virus Activation/physiology , Virus Latency/physiology , Animals , B-Cell Activating Factor/pharmacology , Cyclin-Dependent Kinase Inhibitor p18/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p18/deficiency , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cyclins/pharmacology , DNA Primers/genetics , Flow Cytometry , Gammaherpesvirinae/genetics , Immunoblotting , Mice , Neutralization Tests , Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Virus Activation/drug effects
7.
Stem Cells Dev ; 21(10): 1597-603, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-21936707

ABSTRACT

Cyclin dependent kinase inhibitors (CDKIs) influence proliferation of hematopoietic progenitor cells (HPCs), but little is known of how they influence proliferative responsiveness of HPCs to colony stimulating factors (CSFs), alone and in combination with other hematopoietically active factors, such as the potent co-stimulating cytokine stem cell factor (SCF), or inhibition by myelosuppressive chemokines. Using mice with deletions in p18(INK4c), p21(CIP1/WAF1), or p27(KIP1) genes, and in mice with double gene deletions for either p18/p21 or p18/p27, we determined effects of absence of these CDKIs and their interactions on functional HPC numbers in vivo, and HPC proliferative responsiveness in vitro. There is a decrease in bone marrow HPC proliferation in p18(-/-) mice commensurate with decreased numbers of HPC, suggesting a positive role for p18 on HPC in vivo, similar to that for p21. These positive effects of p18 dominate negative effects of p27 gene deletion. Moreover, the CDKIs differentially regulate responsiveness of granulocyte macrophage (GM) progenitors to synergistic cell proliferation in response to GM-CSF plus SCF, which is considered important for normal hematopoiesis. Responsiveness of HPCs to inhibition by myelosuppressive chemokines is directly related to the capacity of HPCs to respond to synergistic stimulation, and their cell cycle status. P18(INK4c) gene deletion rescued the loss of chemokine suppression of synergistic proliferation due to deletion of p21(CIP1/WAF1). These findings underscore the complex interplay of cell cycle regulators in HPC, and demonstrate that loss of one can sometimes be compensated by loss of another CDKI in both, a pro- or anti-proliferative context.


Subject(s)
Cyclin-Dependent Kinase Inhibitor Proteins/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Hematopoietic Stem Cells/physiology , Stem Cell Factor/physiology , Animals , Bone Marrow Cells/physiology , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Cytokines/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Progenitor Cells/physiology , Intercellular Signaling Peptides and Proteins/physiology , Interleukin-6/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , S Phase , Spleen/cytology , Stem Cell Factor/pharmacology
8.
Mol Oncol ; 5(1): 24-35, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21112821

ABSTRACT

Inherited activating mutations in RET predispose humans to Multiple Endocrine Neoplasia type-2 (MEN2). The MEN2A-specific mutation RET(C634R), RET2A, has been shown to simultaneously downregulate the CDKIs p18 and p27, and upregulate cyclin D1. Importantly, the loss of p18 is necessary and sufficient for RET2A-mediated hyperproliferation. The loss of N-Myc in mice results in embryonic lethality due to a lack of neuronal progenitor cells that fail to proliferate, correlate with accumulation of p18 and p27. Therefore, N-Myc may regulate expression of both CDKIs. Also, N-Myc is expressed predominantly in neuroendocrine cells that give rise to the primary cell types affected in MEN2A. Together these studies suggest that N-Myc is a downstream target of RET2A signaling that prevents accumulation of p18 and/or p27. We report that MAPK activation by RET2A leads to a transient induction of N-Myc mRNA and protein levels, and that N-Myc induction is required to maintain low p18 and p27 levels. Induced N-Myc levels correlate with increased binding of N-Myc to an initiator consensus binding site in the p18 promoter, and this binding is essential for RET2A-mediated transcriptional regulation of p18. Finally, loss of N-Myc induction prevents RET2A-mediated hyperproliferation. Our results demonstrate for the first time that N-Myc is a downstream target of RET2A signaling, and propose that induction of N-Myc by RET2A is a key step leading to lower p18 levels during MEN2A tumorigenesis.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p18/genetics , Gene Expression Regulation/physiology , Proto-Oncogene Proteins c-myc/physiology , Proto-Oncogene Proteins c-ret/metabolism , Signal Transduction , Transcription, Genetic/physiology , Humans , Promoter Regions, Genetic
9.
Cancer Res ; 68(5): 1329-37, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18316595

ABSTRACT

Activating mutations in the RET proto-oncogene are associated with both familial and sporadic medullary thyroid carcinoma (MTC) development; however, the genetic mechanisms underlying MTC tumorigenesis remain largely unknown. Recently, we have identified somatic inactivating mutations in the cell cycle inhibitor gene P18 in human MTC, which coincided with activating RET mutations, suggesting a role for loss of P18 in combination with oncogenic RET in the multistep process of MTC development. Therefore, we crossed transgenic mice expressing oncogenic RET (RET2B) with mice lacking p18 (and p27, another cell cycle inhibitor) and monitored MTC development. RET2B;p18(+/-) mice and RET2B;p18(-/-) mice developed MTC with a highly increased incidence compared with their corresponding single mutant littermates. In addition, expression of oncogenic RET causes an earlier age of onset and larger MTCs in p18(-/-);p27(+/-) mice. In a subset of MTCs of RET2B;p18(+/-)(;p27(+/-)) mice, p18(Ink4c) expression was completely lost. This loss of p18(Ink4c) expression correlated with higher proliferation rates as well as with larger MTCs, indicating that loss of p18 in combination with oncogenic RET not only increases the risk for MTC development but also enhances MTC progression. Our data strongly indicate that oncogenic RET and loss of p18 cooperate in the multistep tumorigenesis of MTC.


Subject(s)
Carcinoma, Medullary/genetics , Carcinoma, Medullary/pathology , Cyclin-Dependent Kinase Inhibitor p18/metabolism , Gene Expression Regulation, Neoplastic , Mutation , Proto-Oncogene Proteins c-ret/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Animals , Calcitonin/metabolism , Cell Cycle , Cell Transformation, Neoplastic , Disease Progression , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Mas
10.
Mol Cell Biol ; 26(23): 8826-39, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16982676

ABSTRACT

Mechanisms coordinating neural progenitor cell cycle exit and differentiation are incompletely understood. The cyclin-dependent kinase inhibitor p27(Kip1) is transcriptionally induced, switching specific neural progenitors from proliferation to differentiation. However, neuronal differentiation-specific transcription factors mediating p27(Kip1) transcription have not been identified. We demonstrate the homeodomain transcription factor Phox2a, required for central nervous system (CNS)- and neural crest (NC)-derived noradrenergic neuron differentiation, coordinates cell cycle exit and differentiation by inducing p27(Kip1) transcription. Phox2a transcription and activation in the CNS-derived CAD cell line and primary NC cells is mediated by combined cyclic AMP (cAMP) and bone morphogenetic protein 2 (BMP2) signaling. In the CAD cellular model, cAMP and BMP2 signaling initially induces proliferation of the undifferentiated precursors, followed by p27(Kip1) transcription, G(1) arrest, and neuronal differentiation. Small interfering RNA silencing of either Phox2a or p27(Kip1) suppresses p27(Kip1) transcription and neuronal differentiation, suggesting a causal link between p27(Kip1) expression and differentiation. Conversely, ectopic Phox2a expression via the Tet-off expression system promotes accelerated CAD cell neuronal differentiation and p27(Kip1) transcription only in the presence of cAMP signaling. Importantly, endogenous or ectopically expressed Phox2a activated by cAMP signaling binds homeodomain cis-acting elements of the p27(Kip1) promoter in vivo and mediates p27(Kip1)-luciferase expression in CAD and NC cells. We conclude that developmental cues of cAMP signaling causally link Phox2a activation with p27(Kip1) transcription, thereby coordinating neural progenitor cell cycle exit and differentiation.


Subject(s)
Cell Cycle/physiology , Cell Differentiation/physiology , Cyclic AMP/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Homeodomain Proteins/genetics , Neural Crest/embryology , Stem Cells/cytology , Transcription, Genetic , Animals , Cell Line , Cells, Cultured , Chromatin Immunoprecipitation , Coturnix/embryology , Cyclic AMP/physiology , Immunohistochemistry , Models, Biological , Neural Crest/cytology , Neural Crest/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Transfection
11.
Mol Carcinog ; 42(2): 109-20, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15584024

ABSTRACT

Mice lacking both p18(Ink4c) and p27(Kip1) develop a tumor spectrum similar to pRb(+/-) mice, and loss of p53 function accelerates tumorigenesis in pRb(+/-) mice. We hypothesized that codeletion of either p18 or p27 in conjunction with p53 deletion will also accelerate tumorigenesis. Mice lacking both p18 and p53 develop several tumors not reported in either single null genotype, including hepatocellular carcinoma, testicular choriocarcinoma, hemangiosarcoma, leiomyosarcoma, fibrosarcoma, and osteosarcoma. Mice lacking both p27 and p53 exhibit a decreased lifespan and develop unique tumors, including papillary carcinoma of the colon, hemangiosarcoma, and leiomyosarcoma. In both p18/p53 and p27/p53 double null genotypes, the incidence and spectra of tissues that develop lymphoma are also increased, as compared to the single null genotypes. The development of p27/p53 double null colon tumors correlates with secondary changes in cell-cycle protein expression and CDK (cyclin-dependent kinase) activity, perhaps contributing to the progression of colorectal cancer. We concluded that p18 and p27 can, not only functionally collaborate with one another, but also can independently collaborate with p53 to modulate the cell cycle and suppress tumorigenesis in a tissue-specific manner.


Subject(s)
Cell Cycle Proteins/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Animals , Blotting, Western , Cell Cycle , Cell Cycle Proteins/biosynthesis , Crosses, Genetic , Cyclin-Dependent Kinase Inhibitor p18 , Cyclin-Dependent Kinase Inhibitor p27 , Disease Progression , Gene Deletion , Genotype , Immunohistochemistry , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Phenotype , Polymerase Chain Reaction , Time Factors , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Proteins/biosynthesis
12.
Exp Cell Res ; 300(2): 365-78, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15475001

ABSTRACT

Myogenic differentiation is characterized by permanent and irreversible cell cycle withdrawal and increased resistance to apoptosis. These functions correlate with changes in expression and activity of several cyclin-dependent kinase inhibitors, including p18, p21, and p27. In this study, we examined the requirements for p18, p21, and p27 in initiating growth arrest in multinucleated myotubes under differentiation conditions and in maintaining terminal arrest upon restimulation of differentiated myotubes with mitogenic signals. Under differentiation conditions, only p27(-/-) or p18(-/-)p27(-/-) myotubes are capable of reentering the cell cycle and synthesizing DNA at a very low frequency. Escape from cell cycle arrest was significantly greater in p18(-/-)p27(-/-) myotubes than in p27(-/-) myotubes. Stimulation of differentiated cultures with a mitogen-rich growth medium enhances p18(-/-)p27(-/-) myotube proliferation to encompass approximately half of the nuclei. p18(-/-)p21(-/-) and p21(-/-)p27(-/-) myotubes remain terminally arrested. Nuclei within individual restimulated p18(-/-)p27(-/-) myotubes can be found in all phases of the cell cycle, and a myotube can be multiphasic without any obvious deleterious effects. Increasing the time of differentiation or serum stimulation of p18(-/-)p27(-/-) myotubes neither increases the proliferation index of the myotube nuclei, nor does it alter the percentage of nuclei in each of the cell cycle phases. During the first 24 h of serum stimulation, the p18(-/-)p27(-/-) myotube nuclei that escape G0 arrest will rearrest in either S or G2 phase, without either mitosis or endoreplication. Apoptosis is increased in restimulated p18(-/-)p27(-/-) myotube nuclei, but is not specific for any cell cycle phase. These results suggest a collaborative role for p18 and p27 in initiating and maintaining G0 arrest during myogenic differentiation. While p18 and p27 appear to be essential in initiating G0 arrest in a proportion of postmitotic myotube nuclei, there must be another cell cycle inhibitor protein that functions with p18 and p27 in maintaining terminal arrest. We propose that the combined rate-limiting expressions of p18, p27, and this other inhibitor determine whether the myotube nuclei will remain postmitotic, or reenter the cell cycle, and if the nuclei escape G0 arrest, in which phase of the cell cycle the nuclei will ultimately rearrest.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Muscle Fibers, Skeletal/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Differentiation/physiology , Cell Division/physiology , Cyclin-Dependent Kinase Inhibitor p18 , Cyclin-Dependent Kinase Inhibitor p27 , Mice , Muscle Fibers, Skeletal/cytology , Myoblasts/cytology , Myoblasts/metabolism , Myosin Heavy Chains/metabolism , Time Factors
13.
Nat Cell Biol ; 6(5): 436-42, 2004 May.
Article in English | MEDLINE | ID: mdl-15122268

ABSTRACT

Self-renewal of stem cells is critical for tissue repair and maintenance of organ integrity in most mammalian systems. The relative asymmetry between self-renewal and differentiation in balance with apoptosis determines the size and durability of a stem-cell pool. Regulation of the cell cycle is one of the fundamental mechanisms underlying determination of cell fate. Absence of p21(Cip1/Waf1), a late G1-phase cyclin-dependent kinase inhibitor (CKI), has previously been shown to enable cell-cycle entry of haematopoietic stem cells, but leads to premature exhaustion of the stem cells under conditions of stress. We show here that deletion of an early G1-phase CKI, p18(INK4C), results in strikingly improved long-term engraftment, largely by increasing self-renewing divisions of the primitive cells in murine transplant models. Therefore, different CKIs have highly distinct effects on the kinetics of stem cells, possibly because of their active position in the cell cycle, and p18(INK4C) appears to be a strong inhibitor limiting the potential of stem-cell self-renewal in vivo.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Division/physiology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Tumor Suppressor Proteins/metabolism , Animals , Bone Marrow Cells/metabolism , Cell Cycle Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p18 , Genotype , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Tumor Suppressor Proteins/genetics
14.
Immunity ; 17(2): 179-89, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12196289

ABSTRACT

B cell terminal differentiation is associated with the onset of high-level antibody secretion and cell cycle arrest. Here the cyclin-dependent kinase (CDK) inhibitor p18(INK4c) is shown to be required within B cells for both terminating cell proliferation and differentiation of functional plasma cells. In its absence, B cells hyperproliferate in germinal centers and extrafollicular foci in response to T-dependent antigens but serum antibody titers are severely reduced, despite unimpaired germinal center formation, class switch recombination, variable region-directed hypermutation, and differentiation to antibody-containing plasmacytoid cells. The novel link between cell cycle control and plasma cell differentiation may, at least in part, relate to p18(INK4c) inhibition of CDK6. Cell cycle arrest mediated by p18(INK4C) is therefore requisite for the generation of functional plasma cells.


Subject(s)
Cell Cycle Proteins , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/immunology , Plasma Cells/cytology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Tumor Suppressor Proteins/immunology , Animals , Antibody Formation , Apoptosis , CD40 Antigens/immunology , Cell Cycle , Cell Differentiation , Cell Division , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinase Inhibitor p18 , Germinal Center , Mice , Mice, Knockout , Plasma Cells/immunology , Signal Transduction/immunology , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...