Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Article in English | MEDLINE | ID: mdl-33574062

ABSTRACT

P27, a cell cycle inhibitor, is also able to drive repression of Sox2 This interaction plays a crucial role during development of p27-/- pituitary tumors because loss of one copy of Sox2 impairs tumorigenesis [H. Li et al., Cell Stem Cell 11, 845-852 (2012)]. However, SOX2 is expressed in both endocrine and stem cells (SCs), and its contribution to tumorigenesis in either cell type is unknown. We have thus explored the cellular origin and mechanisms underlying endocrine tumorigenesis in p27-/- pituitaries. We found that pituitary hyperplasia is associated with reduced cellular differentiation, in parallel with increased levels of SOX2 in stem and endocrine cells. Using conditional loss-of-function and lineage tracing approaches, we show that SOX2 is required cell autonomously in p27-/- endocrine cells for these to give rise to tumors, and in SCs for promotion of tumorigenesis. This is supported by studies deleting the Sox2 regulatory region 2 (Srr2), the target of P27 repressive action. Single cell transcriptomic analysis further reveals that activation of a SOX2-dependent MAPK pathway in SCs is important for tumorigenesis. Altogether, our data highlight different aspects of the role of SOX2 following loss of p27, according to cellular context, and uncover an unexpected SOX2-dependent tumor-promoting role for SCs. Our results imply that targeting SCs, in addition to tumor cells, may represent an efficient antitumoral strategy in certain contexts.


Subject(s)
Carcinogenesis/metabolism , Pituitary Neoplasms/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Carcinogenesis/genetics , Cell Lineage , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cyclin-Dependent Kinase Inhibitor p27/genetics , Endocrine Cells/metabolism , Loss of Function Mutation , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Neoplastic Stem Cells/metabolism , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Protein Domains , SOXB1 Transcription Factors/chemistry , SOXB1 Transcription Factors/genetics
2.
Cell Death Dis ; 10(4): 271, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894510

ABSTRACT

Mechanisms underlying the generation of induced pluripotent stem cells (iPSC) and keeping iPSC stability remain to be further defined. Accumulated evidences showed that iPSC reprogramming may be controlled by the cell-division-rate-dependent model. Here we reported effects of absence of mouse p27 or p18 on iPSC generation efficiency and genomic stability. Expression levels of cyclin-dependent kinases inhibitors (CDKIs), p21, p27, and p18 decreased during iPSC reprogramming. Like p21 loss, p27 or p18 deficiency significantly promoted efficiency of iPSC generation, whereas ectopic expression of p27, p18, or treatment with CDK2 or CDK4 inhibitors repressed the reprogramming rate, suggesting that CDKIs-regulated iPSC reprogramming is directly related with their functions as CDK inhibitors. However, unlike p21 deletion, absence of p27 or p18 did not increase DNA damage or chromosomal aberrations during iPSC reprogramming and at iPSC stage. Our data not only support that cell cycle regulation is critical for iPSC reprogramming, but also reveal the distinction of CDKIs in somatic cell reprogramming.


Subject(s)
Cellular Reprogramming/genetics , Cyclin-Dependent Kinase Inhibitor p18/deficiency , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Genomic Instability/genetics , Induced Pluripotent Stem Cells/metabolism , Animals , Cell Division/genetics , Chromosome Aberrations , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p18/genetics , Cyclin-Dependent Kinase Inhibitor p18/metabolism , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Fibroblasts/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Transduction, Genetic
3.
J Pathol ; 247(1): 60-71, 2019 01.
Article in English | MEDLINE | ID: mdl-30206932

ABSTRACT

The cell cycle inhibitor p27Kip1 is a tumor suppressor via the inhibition of CDK complexes in the nucleus. However, p27 also plays other functions in the cell and may acquire oncogenic roles when located in the cytoplasm. Activation of oncogenic pathways such as Ras or PI3K/AKT causes the relocalization of p27 in the cytoplasm, where it can promote tumorigenesis by unclear mechanisms. Here, we investigated how cytoplasmic p27 participates in the development of non-small cell lung carcinomas. We provide molecular and genetic evidence that the oncogenic role of p27 is mediated, at least in part, by binding to and inhibiting the GTPase RhoB, which normally acts as a tumor suppressor in the lung. Genetically modified mice revealed that RhoB expression is preferentially lost in tumors in which p27 is absent and maintained in tumors expressing wild-type p27 or p27CK- , a mutant that cannot inhibit CDKs. Moreover, although the absence of RhoB promoted tumorigenesis in p27-/- animals, it had no effect in p27CK- knock-in mice, suggesting that cytoplasmic p27 may act as an oncogene, at least in part, by inhibiting the activity of RhoB. Finally, in a cohort of lung cancer patients, we identified a subset of tumors harboring cytoplasmic p27 in which RhoB expression is maintained and these characteristics were strongly associated with decreased patient survival. Thus, monitoring p27 localization and RhoB levels in non-small cell lung carcinoma patients appears to be a powerful prognostic marker for these tumors. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Adenocarcinoma of Lung/enzymology , Carcinoma, Non-Small-Cell Lung/enzymology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cytoplasm/enzymology , Lung Neoplasms/enzymology , rhoB GTP-Binding Protein/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Animals , Carcinogenesis , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cytoplasm/genetics , Cytoplasm/pathology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, 129 Strain , Mice, Knockout , Protein Binding , Signal Transduction , rhoB GTP-Binding Protein/genetics
4.
Cell Cycle ; 17(5): 580-588, 2018.
Article in English | MEDLINE | ID: mdl-29157111

ABSTRACT

We have previously reported a critical role of HMGA proteins in pituitary tumorigenesis since either the Hmga1 or Hmga2 gene overexpression/activation induces the development of mixed growth hormone/prolactin cell pituitary adenomas by activating the E2F transcription factor 1, and then enhancing the G1/S transition of the cell cycle. Consistently, amplification and overexpression of the HMGA2 gene was found in human pituitary prolactinomas. Since impairment of the cell cycle control represents a feature of experimental and human pituitary adenomas, we have investigated the possible synergism between the alterations of other cell cycle regulators, such as p27 deficiency or Cdk4R24C mutation, with Hmga2 overexpression in pituitary tumorigenesis. Therefore, we crossed the Hmga2/T mice, overexpressing the truncated/active form of the Hmga2 gene, either with the knockout mice for p27kip1, or with the knockin mice for the Cdk4R24C mutation, both developing pituitary adenomas. Increased incidence and decreased latency in the development of pituitary lesions appeared in double mutant Hmga2/T;Cdk4R24C mice, and increased features of invasiveness and atypia were observed in pituitary tumors of both Hmga2/T;p27-ko and Hmga2/T;Cdk4R24C double mutant mice as compared with single mutant compounds. Interestingly, most of these mice develop pituitary adenomas with high Ki67 index, extrasellar expansion and brain tissue infiltration, representing good mouse models for human aggressive pituitary adenomas. Taken together, the results reported here indicate a cooperation between HMGA2 overexpression and either p27kip1 or CDK4 impairment in promoting pituitary tumor development and progression.


Subject(s)
Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , HMGA2 Protein/genetics , Pituitary Neoplasms/pathology , Animals , Cell Proliferation , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Disease Models, Animal , Disease-Free Survival , Female , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/mortality
5.
Eur J Immunol ; 47(7): 1142-1152, 2017 07.
Article in English | MEDLINE | ID: mdl-28471480

ABSTRACT

T-cell development is a spatially and temporally regulated process, orchestrated by well-defined contributions of transcription factors and cytokines. Here, we identify the noncoding RNA miR-142 as an additional regulatory layer within murine thymocyte development and proliferation. MiR-142 deficiency impairs the expression of cell cycle-promoting genes in mature mouse thymocytes and early progenitors, accompanied with increased levels of cyclin-dependent kinase inhibitor 1B (Cdkn1b, also known as p27Kip1 ). By using CRISPR/Cas9 technology to delete the miR-142-3p recognition element in the 3'UTR of cdkn1b, we confirm that this gene is a novel target of miR-142-3p in vivo. Increased Cdkn1b protein expression alone however was insufficient to cause proliferation defects in thymocytes, indicating the existence of additional critical miR-142 targets. Collectively, we establish a key role for miR-142 in the control of early and mature thymocyte proliferation, demonstrating the multifaceted role of a single miRNA on several target genes.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/genetics , MicroRNAs/metabolism , Thymocytes/physiology , 3' Untranslated Regions , Animals , CRISPR-Cas Systems , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gene Expression Regulation, Neoplastic , Mice , MicroRNAs/genetics , RNA Processing, Post-Transcriptional
6.
Sci Rep ; 7(1): 595, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28377607

ABSTRACT

Genomic instability represents a typical feature of aggressive cancers. Normal cells have evolved intricate responses to preserve genomic integrity in response to stress, such as DNA damage induced by γ-irradiation. Cyclin-dependent kinases (CDKs) take crucial part to these safeguard mechanisms, but involvement of CDK-inhibitors, such as p27Kip1, is less clear. We generated immortalized fibroblasts from p27kip1 knock-out (KO) mouse embryos and re-expressed p27kip1 WT, or its mutant forms, to identify the function of different domains. We γ-irradiated fibroblasts and observed that loss of p27Kip1 was associated to accumulation of residual DNA damage, increased number of mitotic aberration and, eventually, to survival advantage. Nuclear localization and cyclin/CDK-binding of p27Kip1 were critical to mediate proper response to DNA damage. In human luminal breast cancer (LBC) p27kip1 is frequently down-modulated and CDKN1B, p27Kip1 gene, sporadically mutated. We recapitulated results obtained in mouse fibroblasts in a LBC cell line genetically manipulated to be KO for CDKN1B gene. Following γ-irradiation, we confirmed that p27kip1 expression was necessary to preserve genomic integrity and to recognize and clear-out aberrant cells. Our study provides important insights into mechanisms underlying radio-resistance and unveils the possibility for novel treatment options exploiting DNA repair defects in LBC.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/deficiency , Genomic Instability , Radiation Tolerance/genetics , Animals , Breast Neoplasms , Cell Line, Tumor , Cell Survival/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , DNA Damage/radiation effects , Female , Gene Expression , Gene Knockout Techniques , Humans , MCF-7 Cells , Mice , Micronuclei, Chromosome-Defective , Mitosis/genetics , Mitosis/radiation effects , Mutation , NIH 3T3 Cells
7.
Oncotarget ; 7(43): 69136-69148, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27655701

ABSTRACT

OBJECTIVE: Chronic Helicobacter pylori (H. pylori) infection promotes non-cardia gastric cancer. Some mouse models suggest that bone marrow derived cells (BMDC) contribute to Helicobacter-associated gastric carcinogenesis. We determined whether this increased susceptibility to Helicobacter-induced gastric carcinogenesis of p27-deficient mice is dependent upon their p27-null BMDC or their p27-null gastric epithelial cells. DESIGN: Female mice (recipients) were irradiated and transplanted with BMDC from male donors. Wild type (WT) mice in group 1 (control) received BMDC from male GFP-transgenic mice. Female WT and p27 KO mice were engrafted with male p27KO mice BMDC (Group 2) or GFP-transgenic WT BMDC (Group 3). Recipients were infected with H. pylori SS1 for one year. RESULTS: Mice lacking p27 in either the BM pool or gastric epithelium developed significantly more advanced gastric pathology, including high-grade dysplasia. Co-staining of donor BMDC in dysplastic gastric glands was confirmed by immunofluorescence. Gastric expression of IL-1 beta protein was reduced in groups 2 and 3 (p < 0.05 vs control) whereas expression of IFN-γ and chemokines MIP-1 beta, MIG, IP-10 and RANTES in group 2 were significantly higher than group 3. CONCLUSIONS: Both bone marrow-derived and gastric epithelial cells contribute to the increased gastric cancer susceptibility of p27-deficient H. pylori-infected mice.


Subject(s)
Bone Marrow Cells/metabolism , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Helicobacter Infections/metabolism , Stomach Neoplasms/metabolism , Animals , Bone Marrow Cells/microbiology , Bone Marrow Transplantation/methods , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cytokines/metabolism , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/pathology , Female , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Helicobacter Infections/microbiology , Helicobacter pylori/physiology , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mosaicism , Stomach Neoplasms/microbiology
8.
Circ J ; 80(8): 1795-803, 2016 Jul 25.
Article in English | MEDLINE | ID: mdl-27301409

ABSTRACT

BACKGROUND: Ventricular non-compaction is characterized by a thin layer of compact ventricular myocardium and it is an important abnormality in the mouse heart. It is reminiscent of left ventricular non-compaction, a fairly common human congenital cardiomyopathy. Non-compaction in transgenic mice has been classically evaluated by measuring the thickness of the compact myocardium through histological techniques involving image analysis of 2-dimensional (D) sections. Given the 3D nature of the heart, the aim of this study was to determine whether a technique for the non-destructive, 3D assessment of the mouse embryonic compact myocardium could be developed. METHODS AND RESULTS: Micro-computed tomography (micro-CT), in combination with iodine staining, enabled the differentiation of the trabecular from the compact myocardium in wild-type mice. The 3D and digital nature of the micro-CT data allowed computation anatomical techniques to be readily applied, which were demonstrated via construction of group atlases and atlas-based descriptive statistics. Finally, micro-CT was used to identify the presence of non-compaction in mice with a deletion of the cell cycle inhibitor protein, p27(Kip1). CONCLUSIONS: Iodine staining-enhanced micro-CT with computational anatomical analysis represents a valid addition to classical histology for the delineation of compact myocardial wall thickness in the mouse embryo. Given the quantitative 3D resolution of micro-CT, these approaches might provide helpful information for the analysis of non-compaction. (Circ J 2016; 80: 1795-1803).


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/deficiency , Embryo, Mammalian , Heart Defects, Congenital , Myocardium , X-Ray Microtomography , Animals , Embryo, Mammalian/diagnostic imaging , Embryo, Mammalian/embryology , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Humans , Mice , Mice, Knockout
9.
Microvasc Res ; 103: 36-40, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26522285

ABSTRACT

The proliferation of vascular smooth muscle cells (SMCs) contributes to atherosclerotic plaque formation and restenosis. Cyclin-dependent kinase inhibitors, such as p27(Kip1) and p21(Cip1), are known to play significant roles in the control of the aberrant proliferation of SMCs. Primary cultured SMCs stop proliferating immediately when cultured in three-dimensional matrices of type-I collagen "honeycombs" structures. To clarify whether p27(Kip1) and p21(Cip1) are involved in the proliferative inhibition of SMCs cultured in honeycombs, the characteristics of SMCs derived from the aorta of both wild-type mice (p27[+/+] SMCs) and p27(Kip1) knockout mice (p27[-/-] SMCs) were investigated. Although the growth of p27(-/-) SMCs cultured on plates was faster than that of p27(+/+) SMCs, the number of both p27(+/+) and p27(-/-) SMCs did not change when they were cultured in honeycombs. p21(Cip1) expression was decreased but maintained in p27(-/-) SMCs cultured on plates and in honeycombs. Knockdown of p21(Cip1) in p27(-/-) SMCs promoted proliferation on plates. On the contrary, p21(Cip1) knockdown had no effect on the proliferation of p27(-/-) SMCs cultured in honeycombs. In conclusion, p27(Kip1) and p21(Cip1) are insufficient for the proliferative inhibition of SMCs cultured in honeycombs.


Subject(s)
Cell Proliferation , Collagen Type I/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Cells, Cultured , Collagen Type I/chemistry , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cyclin-Dependent Kinase Inhibitor p27/genetics , Female , Gene Expression Regulation , Genotype , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Phenotype , Porosity , Primary Cell Culture , RNA Interference , Signal Transduction , Time Factors , Transfection
10.
J Vasc Surg ; 63(5): 1351-9, 2016 05.
Article in English | MEDLINE | ID: mdl-25701497

ABSTRACT

OBJECTIVE: The natural response to arterial occlusive disease is enlargement of collaterals; however, the molecular factors that control collateralization are not well understood. The gene p27(Kip1) (p27) affects human response to arterial injury. Previous studies have shown that overexpression of p27 inhibits vascular endothelial and vascular smooth muscle cell (VSMC) proliferation and angiogenesis. To test the hypothesis that knockout of p27 would improve collateralization in reaction to ischemia, we performed in vivo and in vitro experiments using p27 knockout (p27(-/-)) and wild-type (wt) mice. METHODS: Hindlimb ischemia was induced by left femoral artery ligation in p27(-/-) and wt (C57BL/6) female mice. The mice underwent weekly laser Doppler perfusion imaging of the footpads until sacrifice on postoperative day 28 followed by microcomputed tomography scanning of both hindlimbs. VSMCs were isolated from p27(-/-) and wt mice and used in migration and gel contraction assays in the absence and presence of the nonspecific matrix metalloproteinase (MMP) inhibitor BB94. MMP-2 and MMP-9 messenger RNA (mRNA) expression was measured by quantitative reverse transcription-polymerase chain reaction in p27(-/-) and wt VSMCs. RESULTS: p27(-/-) mice reperfused more effectively than wt mice by laser Doppler starting from day 7 (ischemic/nonischemic ratio, 0.33 ± 0.02 vs 0.25 ± 0.02; P < .05) and continuing through day 28 (0.45 ± 0.04 vs 0.31 ± 0.04; P < .05). The gracilis collateral diameter was similar for the nonischemic hindlimbs of the p27(-/-) and wt mice, and this collateral pathway increased similarly after ischemia as assessed by microcomputed tomography. However, the p27(-/-) mice significantly enlarged a novel collateral pathway that bridged directly between the femoral artery proximal to the ligation site and the saphenous or popliteal artery distal to the ligation site more than wt mice (158 ± 18.3 vs 82 ± 22 µm; P < .001). p27(-/-) VSMCs migrated more (79% ± 5% vs 56% ± 6%; P < .05) and caused more gel contraction (18% ± 5% of the initial area vs 43% ± 4%; P < .05) than wt cells. Migration and collagen contraction were abolished in p27(-/-) and wt cells by MMP inhibition. p27(-/-) cells expressed significantly more MMP-2 mRNA than wt cells did. CONCLUSIONS: Knockout of p27 enhances arterial collateralization in response to hindlimb ischemia through enlargement of a new collateral pathway. In vitro, knockout of p27 increases collagen gel contraction in addition to stimulating VSMC migration. We speculate that p27 may affect collateralization through its role in regulating MMP-2 expression.


Subject(s)
Collateral Circulation , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Ischemia/physiopathology , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Angiogenesis Inhibitors/pharmacology , Animals , Blood Flow Velocity , Cell Movement , Cells, Cultured , Collagen/metabolism , Collateral Circulation/drug effects , Cyclin-Dependent Kinase Inhibitor p27/genetics , Disease Models, Animal , Female , Genotype , Hindlimb , Ischemia/genetics , Ischemia/metabolism , Laser-Doppler Flowmetry , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic/drug effects , Phenotype , Regional Blood Flow , Time Factors , X-Ray Microtomography
11.
Oncotarget ; 6(34): 35880-92, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26416424

ABSTRACT

p27Kip1 (p27) is a negative regulator of proliferation and a tumor suppressor via the inhibition of cyclin-CDK activity in the nucleus. p27 is also involved in the regulation of other cellular processes, including transcription by acting as a transcriptional co-repressor. Loss of p27 expression is frequently observed in pancreatic adenocarcinomas in human and is associated with decreased patient survival. Similarly, in a mouse model of K-Ras-driven pancreatic cancer, loss of p27 accelerates tumor development and shortens survival, suggesting an important role for p27 in pancreatic tumorigenesis. Here, we sought to determine how p27 might contribute to early events leading to tumor development in the pancreas. We found that K-Ras activation in the pancreas causes p27 mislocalization at pre-neoplastic stages. Moreover, loss of p27 or expression of a mutant p27 that does not bind cyclin-CDKs causes the mislocalization of several acinar polarity markers associated with metaplasia and induces the nuclear expression of Sox9 and Pdx1 two transcription factors involved in acinar-to-ductal metaplasia. Finally, we found that p27 directly represses transcription of Sox9, but not that of Pdx1. Thus, our results suggest that K-Ras activation, the earliest known event in pancreatic carcinogenesis, may cause loss of nuclear p27 expression which results in derepression of Sox9, triggering reprogramming of acinar cells and metaplasia.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/deficiency , Pancreas/metabolism , Pancreas/pathology , SOX9 Transcription Factor/biosynthesis , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , HEK293 Cells , HeLa Cells , Humans , Metaplasia , Mice , Mice, Knockout , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Signal Transduction
12.
Oncotarget ; 6(26): 23026-35, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26246476

ABSTRACT

Primary melanoma, a highly aggressive malignancy, exhibits heterogeneity in biologic behaviors, clinical characteristics, metastasis potential and mortality. The present study sought to identify the molecular signatures that define a subgroup of primary melanomas with high risks of metastasis and mortality. First, we identified the markers that best differentiated metastatic melanomas from primary melanomas by examining the expression of seven previously reported biomarkers (BRAF, Dicer, Fbw7, KAI1, MMP2, p27 and Tip60) in a training cohort consisting of 145 primary melanomas and 105 metastatic melanomas. KAI1 and p27, both tumor suppressors, emerged as best candidates. Loss of both tumor suppressors occurred in the majority (74.29%) of metastatic melanomas. Further, a subset (metastatic like, or "ML", 33.10%) of primary melanomas also lost these two tumor suppressors. Kaplan-Meier analysis indicated that ML subgroup of primary melanoma patients had much worse 5 year survival compared with other primary melanoma patients (P = 0.002). The result was confirmed in an independent validation cohort with 92 primary melanomas (P = 0.030) and in the combined cohort with 237 melanoma patients (P = 3.00E-4). Additionally, compared to KAI1 and p27 as an individual prognostic marker, the combined signature is more closely associated with melanoma patient survival (P = 0.025, 0.264 and 0.009, respectively). In conclusion, loss of both KAI1 and p27 defines a subgroup of primary melanoma patients with poor prognosis. This molecular signature may help in metastatic melanoma diagnosis and may provide information useful in identifying high-risk primary melanoma patients for more intensive clinical surveillance in the future.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/deficiency , Kangai-1 Protein/deficiency , Melanoma/classification , Melanoma/metabolism , Skin Neoplasms/genetics , Biomarkers, Tumor/deficiency , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Humans , Kangai-1 Protein/genetics , Kangai-1 Protein/metabolism , Male , Melanoma/genetics , Melanoma/pathology , Middle Aged , Prognosis , Skin Neoplasms/pathology , Melanoma, Cutaneous Malignant
13.
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
14.
Biol Reprod ; 92(3): 77, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25609837

ABSTRACT

Cyclin-dependent kinase inhibitors p21(Cip1) (CDKN1A) and p27(Kip1) (CDKN1B) are expressed in Leydig cells. Previously, we reported that Cdkn1b knockout in the mouse led to increased Leydig cell proliferative capacity and lower steroidogenesis. However, the relative importance of CDKN1A and CDKN1B in these regulations was unclear. In the present study, we examined the relative importance of CDKN1A and CDKN1B in regulation of Leydig cell proliferation and steroidogenesis by whole-body knockout of CDKN1A (Cdkn1a(-/-)) and CDKN1A/CDKN1B double knockout (DBKO). The cell number, 5-bromo-2-deoxyuridine incorporation rate, steroidogenesis, and steroidogenic enzyme mRNA levels and activities of Leydig cells were compared among wild-type (WT), Cdkn1a(-/-), and DBKO mice. Relative to WT mice, Leydig cell number per testis was doubled in the DBKO and unchanged in the Cdkn1a(-/-) mice. Testicular testosterone levels and mRNA levels for luteinizing hormone receptor (Lhcgr), steroidogenic acute regulatory protein (Star), cholesterol side-chain cleavage enzyme (Cyp11a1), 17alpha-hydroxylase/17,20-lyase (Cyp17a1), and 17beta-hydroxysteroid dehydrogenase 3 (Hsd17b3) and their respective proteins were significantly lower in the DBKO mice. However, testicular testosterone level was unchanged in the Cdkn1a(-/-) mice, although Lhcgr mRNA levels were significantly lower relative to those in the WT control. We conclude that Cdkn1a(-/-) did not increase Leydig cell numbers (although a defect of Leydig cell function was noted), whereas DBKO caused a significant increase of Leydig cell numbers but a decrease of steroidogenesis.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Leydig Cells/cytology , Sexual Maturation/physiology , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/physiology , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/physiology , Leydig Cells/metabolism , Male , Mice , Mice, Knockout , Models, Animal , Phosphoproteins/metabolism , RNA, Messenger/metabolism , Receptors, LH/metabolism , Sexual Maturation/genetics , Steroids/metabolism
15.
Cell Cycle ; 13(19): 3100-11, 2014.
Article in English | MEDLINE | ID: mdl-25486569

ABSTRACT

The CDK inhibitor p27(kip1) is a critical regulator of cell cycle progression, but the mechanisms by which p27(kip1) controls cell proliferation in vivo are still not fully elucidated. We recently demonstrated that the microtubule destabilizing protein stathmin is a relevant p27(kip1) binding partner. To get more insights into the in vivo significance of this interaction, we generated p27(kip1) and stathmin double knock-out (DKO) mice. Interestingly, thorough characterization of DKO mice demonstrated that most of the phenotypes of p27(kip1) null mice linked to the hyper-proliferative behavior, such as the increased body and organ weight, the outgrowth of the retina basal layer and the development of pituitary adenomas, were reverted by co-ablation of stathmin. In vivo analyses showed a reduced proliferation rate in DKO compared to p27(kip1) null mice, linked, at molecular level, to decreased kinase activity of CDK4/6, rather than of CDK1 and CDK2. Gene expression profiling of mouse thymuses confirmed the phenotypes observed in vivo, showing that DKO clustered with WT more than with p27 knock-out tissue. Taken together, our results demonstrate that stathmin cooperates with p27(kip1) to control the early phase of G1 to S phase transition and that this function may be of particular relevance in the context of tumor progression.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/genetics , Stathmin/genetics , Animals , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Proliferation , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Female , G1 Phase , Gene Expression Profiling , Gigantism/metabolism , Gigantism/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pituitary Gland/metabolism , Pituitary Gland/pathology , S Phase , Stathmin/deficiency , Thymus Gland/metabolism , Thymus Gland/pathology
16.
Cancer Lett ; 355(1): 106-12, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25218349

ABSTRACT

H. pylori infection causes gastritis, peptic ulcers and gastric cancer. Eradicating H. pylori prevents ulcers, but to what extent this prevents cancer remains unknown, especially if given after intestinal metaplasia has developed. H. pylori infected wild-type (WT) mice do not develop cancer, but mice lacking the tumor suppressor p27 do so, thus providing an experimental model of H. pylori-induced cancer. We infected p27-deficient mice with H. pylori strain SS1 at 6-8 weeks of age. Persistently H. pylori-infected WT C57BL/6 mice served as controls. Mice in the eradication arms received antimicrobial therapy (omeprazole, metronidazole and clarithromycin) either "early" (at 15 weeks post infection, WPI) or "late" at 45 WPI. At 70 WPI, mice were euthanized for H. pylori determination, histopathology and cytokine/chemokine expression. Persistently infected mice developed premalignant lesions including high-grade dysplasia, whereas those given antibiotics did not. Histologic activity scores in the eradication groups were similar to each other, and were significantly decreased compared with controls for inflammation, epithelial defects, hyperplasia, metaplasia, atrophy and dysplasia. IP-10 and MIG levels in groups that received antibiotics were significantly lower than controls. There were no significant differences in expression of IFN-γ, TNF-α, IL-1ß, RANTES, MCP-1, MIP-1α or MIP-1ß among the three groups. Thus, H. pylori eradication given either early or late after infection significantly attenuated gastric inflammation, gastric atrophy, hyperplasia, and dysplasia in the p27-deficient mice model of H. pylori-induced gastric cancer, irrespective of the timing of antibiotic administration. This was associated with reduced expression of IP-10 and MIG.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Gastritis/prevention & control , Helicobacter Infections/drug therapy , Helicobacter pylori/drug effects , Stomach Neoplasms/prevention & control , Stomach/drug effects , Animals , Atrophy , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cytokines/metabolism , Disease Models, Animal , Drug Administration Schedule , Drug Therapy, Combination , Gastric Mucosa/metabolism , Gastritis/genetics , Gastritis/metabolism , Gastritis/microbiology , Gastritis/pathology , Helicobacter Infections/complications , Helicobacter Infections/genetics , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/pathogenicity , Hyperplasia , Inflammation Mediators/metabolism , Metaplasia , Mice, Inbred C57BL , Mice, Knockout , Proton Pump Inhibitors/administration & dosage , Stomach/microbiology , Stomach/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Time Factors
17.
Clin Exp Pharmacol Physiol ; 41(10): 807-16, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24916068

ABSTRACT

To assess the role of p27(kip1) in regulating dental formation and alveolar bone development, we compared the teeth and mandible phenotypes of homozygous p27(kip1) -deficient (p27(-/-) ) mice with their wild-type littermates at 2 weeks of age. At 2 weeks of age, dental mineral density, dental volume and dentin sialoprotein-immunopositive areas were increased significantly, whereas the predentin area : total dentin area and biglycan-immunopositive area : dentin area ratios were decreased significantly in p27(-/-) mice compared with their wild-type (WT) littermates. Mandible mineral density, cortical thickness, alveolar bone volume, type I collagen and osterix-immunopositive areas, osteoblast number and activity and mRNA expression of Runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP), osteocalcin and bone morphogenetic protein (bmp2) were all significantly increased in the mandibles, as was the number and surface of tartrate-resistant acid phosphatase-positive osteoclasts in the alveolar bone of p27(-/-) mice compared with their WT littermates. Furthermore, the percentage of proliferating cell nuclear antigen-positive cells in Hertwig's epithelial root sheath and protein expression of cyclin E and cyclin-dependent kinase 2 were increased significantly in p27(-/-) mice relative to their WT littermates. The results from this study indicate that p27 plays a negative regulatory role in dentin formation and alveolar bone development.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/deficiency , Dentin/physiology , Mandible/physiology , Osteogenesis/physiology , Alkaline Phosphatase/metabolism , Animals , Bone Density/physiology , Bone Morphogenetic Protein 2/metabolism , Collagen Type I/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Dentin/metabolism , Mandible/metabolism , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteoblasts/physiology , Osteocalcin/metabolism , Osteoclasts/metabolism , Osteoclasts/physiology
18.
Biochem Biophys Res Commun ; 447(4): 563-8, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24751519

ABSTRACT

p27(Kip1) is a potent inhibitor of the cyclin-dependent kinases that drive G1 to S phase transition. Since deregulation of p27(Kip1) is found in many malignancies and is associated with the poor prognosis, elucidation of the molecular bases for regulation of p27(Kip1) expression is of great significance, not only in providing insight into the understanding of biological p27(Kip1), but also in the development of new cancer therapeutic tactics. We here explored the inhibitory regulation of IKKß on p27(Kip1) expression following arsenite exposure. We found that although the basal level of p27(Kip1) expression in the IKKß(-/-) cells is much lower than that in the IKKß(+/+) cells, the deletion of IKKß in the MEFs led to a marked increase in p27(Kip1) protein induction due to arsenite exposure in comparison to that in the IKKß(+/+) cells. The IKKß regulatory effect on p27(Kip1) expression was also verified in the IKKß(-/-) and IKKß(-/-) cells with IKKß reconstitutional expression, IKKß(-/-) (IKKß). Further studies indicated that IKKß-mediated p27(Kip1) downregulation occurred at protein degradation level via p65-dependent and p50-independent manner. Moreover, the results obtained from the comparison of arsenite-induced GSK3ß activation among transfectants of WT, IKKß(-/-) and IKKß(-/-) (IKKß), and the utilization of GSKß shRNA, demonstrated that IKKß regulation of p27 protein degradation was mediated by GSK3ß following arsenite exposure.


Subject(s)
Arsenites/pharmacology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , I-kappa B Kinase/metabolism , Transcription Factor RelA/metabolism , Animals , Antineoplastic Agents/pharmacology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cyclin-Dependent Kinase Inhibitor p27/genetics , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Knockout Techniques , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HeLa Cells , Humans , I-kappa B Kinase/deficiency , I-kappa B Kinase/genetics , Mice , NF-kappa B p50 Subunit/deficiency , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , Phosphorylation , Proteolysis/drug effects , RNA, Small Interfering/genetics , Transcription Factor RelA/deficiency , Transcription Factor RelA/genetics
19.
Curr Med Chem ; 20(33): 4131-41, 2013.
Article in English | MEDLINE | ID: mdl-23992309

ABSTRACT

Epidemiological and clinical studies have indicated that low vitamin D activity is not only associated with an increased cancer risk and a more aggressive tumor growth, but also connected with an aggravated liver damage caused by chronic inflammation. Meanwhile, increasing evidence has demonstrated that 1,25(OH)2D3 (the most biologically active metabolite of vitamin D) can inhibit inflammatory response in some chronic inflammatory associated cancer, which is considered to have the anti-tumor potency. However, the interaction between 1,25(OH)2D3 and inflammation during hepatocellular carcinoma (HCC) initiation and progression is not yet clear. Here, we report an anti-tumorigenesis effect of 1,25(OH)2D3 via decreasing inflammatory cytokine secretion in HCC and hypothesize the possible underlying mechanism. Firstly, we show that the enhanced tumor growth is associated with elevated inflammatory cytokine IL-6 and TNF-α in 1α(OH)ase gene-knockout mice. Secondly, 1,25(OH)2D3 can inhibit vitamin D receptor (VDR) shRNA interfered tumor cell growth through decreasing inflammatory cytokine secretion in vitro and in vivo. Finally, using p27(kip1) gene knock-out mouse model, we demonstrate that the effect of 1,25(OH)2D3 in inhibiting immune cell related inflammatory cytokine secretion, exerts in a p27(kip1) gene dependent way. Collectively, 1,25(OH)2D3 inhibits HCC development through up-regulating the expression of p27(kip1) in immune cell and reducing inflammatory cytokine production.


Subject(s)
Calcitriol/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Interleukin-6/metabolism , Liver Neoplasms/drug therapy , Tumor Necrosis Factor-alpha/metabolism , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/deficiency , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Animals , Apoptosis/drug effects , Calcitriol/pharmacology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Calcitriol/antagonists & inhibitors , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transplantation, Homologous , Up-Regulation/drug effects
20.
Development ; 140(15): 3118-27, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23824576

ABSTRACT

Cortical development requires the precise timing of neural precursor cell (NPC) terminal mitosis. Although cell cycle proteins regulate terminal mitosis, the factors that influence the cell cycle machinery are incompletely understood. Here we show in mice that myeloid cell leukemia 1 (Mcl1), an anti-apoptotic Bcl-2 protein required for the survival of NPCs, also regulates their terminal differentiation through the cell cycle regulator p27(Kip1). A BrdU-Ki67 cell profiling assay revealed that in utero electroporation of Mcl1 into NPCs in the embryonic neocortex increased NPC cell cycle exit (the leaving fraction). This was further supported by a decrease in proliferating NPCs (Pax6(+) radial glial cells and Tbr2(+) neural progenitors) and an increase in differentiating cells (Dcx(+) neuroblasts and Tbr1(+) neurons). Similarly, BrdU birth dating demonstrated that Mcl1 promotes premature NPC terminal mitosis giving rise to neurons of the deeper cortical layers, confirming their earlier birthdate. Changes in Mcl1 expression within NPCs caused concomitant changes in the levels of p27(Kip1) protein, a key regulator of NPC differentiation. Furthermore, in the absence of p27(Kip1), Mcl1 failed to induce NPC cell cycle exit, demonstrating that p27(Kip1) is required for Mcl1-mediated NPC terminal mitosis. In summary, we have identified a novel physiological role for anti-apoptotic Mcl1 in regulating NPC terminal differentiation.


Subject(s)
Brain/embryology , Brain/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Neural Stem Cells/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Brain/cytology , Cell Cycle Checkpoints , Cell Differentiation , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cyclin-Dependent Kinase Inhibitor p27/genetics , Doublecortin Protein , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitosis , Myeloid Cell Leukemia Sequence 1 Protein , Neural Stem Cells/cytology , Neurogenesis , Pregnancy , Proto-Oncogene Proteins c-bcl-2/deficiency , Proto-Oncogene Proteins c-bcl-2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...