Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Am J Pathol ; 194(6): 1007-1019, 2024 06.
Article in English | MEDLINE | ID: mdl-38442804

ABSTRACT

Cytotoxic CD8+ T cells are central to the antitumor immune response by releasing cytotoxic granules that kill tumor cells. They are activated by antigen-presenting cells, which become activated by DAMPs (damage associated molecular patterns) through MyD88. However, the suppressive tumor microenvironment promotes T-cell tolerance to tumor antigens, in part by enhancing the activity of immune checkpoint molecules that prevent CD8+ T-cell activation and cytotoxicity. MyD88 limits CD4+ T-cell activation during cardiac adaptation to stress. A similar mechanism is hypothesized to exist in CD8+ T cells that could be modulated to improve antitumor immunity. Herein, adoptive transfer of MyD88-/- CD8+ T cells in melanoma-bearing T-cell-deficient mice resulted in slower tumor growth, greater intratumoral T-cell accumulation, and higher melanoma cell death compared with transfer of wild-type CD8+ T cells. These findings were also observed in T-cell-specific MyD88-/- mice compared with wild-type littermates implanted with melanoma. Mechanistically, deletion of MyD88 enhanced CD8+ T-cell activation and survival, and T-cell receptor induced degranulation of cytotoxic molecules, overall improving the killing of melanoma cells. This enhanced cytotoxicity was retained in mice bearing tumors expressing the specific antigen for which cytotoxic T-cells were restricted. This study's results demonstrate a conserved mechanism for MyD88 in modulating CD8+ T-cell activation and represent a novel target in improving cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Myeloid Differentiation Factor 88 , Animals , Mice , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Melanoma/immunology , Melanoma/pathology , Melanoma/genetics , Melanoma/therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Melanoma, Experimental/genetics , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Tumor Microenvironment/immunology
2.
Proc Natl Acad Sci U S A ; 119(38): e2206147119, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36095192

ABSTRACT

The neocortex, the center for higher brain function, first emerged in mammals and has become massively expanded and folded in humans, constituting almost half the volume of the human brain. Primary microcephaly, a developmental disorder in which the brain is smaller than normal at birth, results mainly from there being fewer neurons in the neocortex because of defects in neural progenitor cells (NPCs). Outer radial glia (oRGs), NPCs that are abundant in gyrencephalic species but rare in lissencephalic species, are thought to play key roles in the expansion and folding of the neocortex. However, how oRGs expand, whether they are necessary for neocortical folding, and whether defects in oRGs cause microcephaly remain important questions in the study of brain development, evolution, and disease. Here, we show that oRG expansion in mice, ferrets, and human cerebral organoids requires cyclin-dependent kinase 6 (CDK6), the mutation of which causes primary microcephaly via an unknown mechanism. In a mouse model in which increased Hedgehog signaling expands oRGs and intermediate progenitor cells and induces neocortical folding, CDK6 loss selectively decreased oRGs and abolished neocortical folding. Remarkably, this function of CDK6 in oRG expansion did not require its kinase activity, was not shared by the highly similar CDK4 and CDK2, and was disrupted by the mutation causing microcephaly. Therefore, our results indicate that CDK6 is conserved to promote oRG expansion, that oRGs are necessary for neocortical folding, and that defects in oRG expansion may cause primary microcephaly.


Subject(s)
Cyclin-Dependent Kinase 6 , Ependymoglial Cells , Microcephaly , Neocortex , Animals , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Ependymoglial Cells/cytology , Ependymoglial Cells/enzymology , Ferrets , Hedgehog Proteins/metabolism , Humans , Mice , Microcephaly/genetics , Neocortex/abnormalities , Neocortex/enzymology , Neural Stem Cells/cytology , Neural Stem Cells/enzymology , Organoids/embryology
3.
Proc Natl Acad Sci U S A ; 117(46): 28806-28815, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33139577

ABSTRACT

Akt activation up-regulates the intracellular levels of reactive oxygen species (ROS) by inhibiting ROS scavenging. Of the Akt isoforms, Akt3 has also been shown to up-regulate ROS by promoting mitochondrial biogenesis. Here, we employ a set of isogenic cell lines that express different Akt isoforms, to show that the most robust inducer of ROS is Akt3. As a result, Akt3-expressing cells activate the DNA damage response pathway, express high levels of p53 and its direct transcriptional target miR-34, and exhibit a proliferation defect, which is rescued by the antioxidant N-acetylcysteine. The importance of the DNA damage response in the inhibition of cell proliferation by Akt3 was confirmed by Akt3 overexpression in p53-/- and INK4a-/-/Arf-/- mouse embryonic fibroblasts (MEFs), which failed to inhibit cell proliferation, despite the induction of high levels of ROS. The induction of ROS by Akt3 is due to the phosphorylation of the NADPH oxidase subunit p47phox, which results in NADPH oxidase activation. Expression of Akt3 in p47phox-/- MEFs failed to induce ROS and to inhibit cell proliferation. Notably, the proliferation defect was rescued by wild-type p47phox, but not by the phosphorylation site mutant of p47phox In agreement with these observations, Akt3 up-regulates p53 in human cancer cell lines, and the expression of Akt3 positively correlates with the levels of p53 in a variety of human tumors. More important, Akt3 alterations correlate with a higher frequency of mutation of p53, suggesting that tumor cells may adapt to high levels of Akt3, by inactivating the DNA damage response.


Subject(s)
DNA Damage , NADPH Oxidases/metabolism , Oxidative Stress/physiology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line , Enzyme Activation , Mice , NADPH Oxidases/genetics , Oxidation-Reduction , Oxidative Stress/genetics , Phosphoproteins/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , Superoxides/metabolism
4.
Eur J Immunol ; 51(11): 2576-2589, 2021 11.
Article in English | MEDLINE | ID: mdl-34363211

ABSTRACT

Regulatory T cells (Tregs) play a critical role in maintaining self-tolerance and controlling inflammation. However, physiologically relevant conditions that alter Treg function and drive disease pathogenesis are poorly understood and few have been defined. We have previously shown that induction of hyperlipidemia in mice results in changes in Tregs that reduce their function. Here, we set out to examine mechanisms by which hyperlipidemia alters Tregs. Using live-cell metabolic assays, we observed that induction of hyperlipidemia increases metabolism in Tregs but not conventional T cells. Increased metabolism resulted from preferential activation of the serine/threonine kinase Akt2 (PKB-ß). Expression of a constitutively activated form of Akt2 in CD4 T cells was sufficient to increase glycolysis in Tregs and drive changes in Treg subsets. Induction of hyperlipidemia did not alter Treg metabolism in mice lacking Akt2. Activation of Akt2 was sufficient to drive the production of inflammatory cytokines by Tregs. We suggest that hyperlipidemia alters Treg function through effects on metabolism via Akt2 activation thereby promoting plasticity and decreased function of FoxP3+ T cells.


Subject(s)
Hyperlipidemias/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Mice , Proto-Oncogene Proteins c-akt/immunology
5.
Am J Physiol Heart Circ Physiol ; 317(7): H26-H36, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30951363

ABSTRACT

The lack of pharmaceutical targets available to treat patients with calcific aortic valve disease (CAVD) necessitates further research into the specific mechanisms of the disease. The significant changes that occur to the aortic valves extracellular matrix (ECM) during the progression of CAVD suggests that these proteins may play an important role in calcification. Exploring the relationship between valve interstitial cells (VICs) and the ECM may lead to a better understand of CAVD mechanisms and potential pharmaceutical targets. In this study, we look at the effect of two ECM components, collagen and hyaluronic acid (HA), on the mineralization of VICs within the context of a two-dimensional, polyacrylamide (PAAM) model system. Using a novel, nondestructive imaging technique, we were able to track calcific nodule development in culture systems over a 3-wk time frame. We saw a significant increase in the size of the nodules grown on HA PAAM gels as compared with collagen PAAM gels, suggesting that HA has a direct effect on mineralization. Directly looking at the two known receptors of HA, CD44 and receptor for HA-mediated motility (RHAMM), and using siRNA knockdown revealed that a decrease in CD44 expression resulted in a reduction of calcification. A decrease in CD44, through siRNA knockdown, reduces mineralization on HA PAAM gels, suggesting a potential new target for CAVD treatment. NEW & NOTEWORTHY Our in vitro model of calcific aortic valve disease shows an interaction between the hyaluronic acid binding protein CD44 with the osteogenic factor OPN as a potential mechanism of aortic valve calcification. Using siRNA knockdown of CD44, we show an upregulation of OPN expression with a decrease in overall mineralization.


Subject(s)
Aortic Valve/metabolism , Calcinosis/genetics , Heart Valve Diseases/genetics , Hyaluronan Receptors/genetics , Animals , Aortic Valve/cytology , Calcinosis/metabolism , Cell Movement , Cells, Cultured , Collagen/metabolism , Extracellular Matrix/metabolism , Heart Valve Diseases/metabolism , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Male , Osteopontin/genetics , Osteopontin/metabolism , Rats , Rats, Sprague-Dawley
6.
Mol Cell ; 44(4): 513-4, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-22099300

ABSTRACT

In this issue of Molecular Cell, Yang et al. (2011) demonstrate that Aurora B phosphorylates ATM, leading to its mitotic activation and ability to phosphorylate Bub1 and regulate the spindle checkpoint, thus maintaining genomic integrity.

7.
Cell Mol Life Sci ; 75(20): 3817-3827, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29728713

ABSTRACT

Neural stem cells give rise to granule dentate neurons throughout life in the hippocampus. Upon activation, these stem cells generate fast proliferating progenitors that complete several rounds of divisions before differentiating into neurons. Although the mechanisms regulating the activation of stem cells have been intensively studied, little attention has been given so far to the intrinsic machinery allowing the expansion of the progenitor pool. The cell cycle protein Cdk6 positively regulates the proliferation of hippocampal progenitors, but the mechanism involved remains elusive. Whereas Cdk6 functions primarily as a cell cycle kinase, it can also act as transcriptional regulator in cancer cells and hematopoietic stem cells. Using mouse genetics, we show here that the function of Cdk6 in hippocampal neurogenesis relies specifically on its kinase activity. The present study also reveals a specific regulatory mechanism for Cdk6 in hippocampal progenitors. In contrast to the classical model of the cell cycle, we observe that the Cip/Kip family member p27, rather than the Ink4 family, negatively regulates Cdk6 in the adult hippocampus. Altogether, our data uncover a unique, cell type-specific regulatory mechanism controlling the expansion of hippocampal progenitors, where Cdk6 kinase activity is modulated by p27.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Animals , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase Inhibitor p18/deficiency , Cyclin-Dependent Kinase Inhibitor p18/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis
8.
Haematologica ; 102(6): 995-1005, 2017 06.
Article in English | MEDLINE | ID: mdl-28255017

ABSTRACT

Mice lacking Cdk6 kinase activity suffer from mild anemia accompanied by elevated numbers of Ter119+ cells in the bone marrow. The animals show hardly any alterations in erythroid development, indicating that Cdk6 is not required for proliferation and maturation of erythroid cells. There is also no difference in stress erythropoiesis following hemolysis in vivo However, Cdk6-/- erythrocytes have a shortened lifespan and are more sensitive to mechanical stress in vitro, suggesting differences in cytoskeletal architecture. Erythroblasts contain both Cdk4 and Cdk6, while mature erythrocytes apparently lack Cdk4 and their Cdk6 is partly associated with the cytoskeleton. We used mass spectrometry to show that Cdk6 interacts with a number of proteins involved in cytoskeleton organization. Cdk6-/- erythroblasts show impaired F-actin formation and lower levels of gelsolin, which interacts with Cdk6. We also found that Cdk6 regulates the transcription of a panel of genes involved in actin (de-)polymerization. Cdk6-deficient cells are sensitive to drugs that interfere with the cytoskeleton, suggesting that our findings are relevant to the treatment of patients with anemia - and may be relevant to cancer patients treated with the new generation of CDK6 inhibitors.


Subject(s)
Cyclin-Dependent Kinase 6/physiology , Cytoskeleton/ultrastructure , Erythroid Cells/ultrastructure , Actin Cytoskeleton , Actins/metabolism , Anemia , Animals , Gelsolin/metabolism , Gene Expression Regulation , Mass Spectrometry , Mice , Mice, Inbred C57BL
9.
J Biol Chem ; 290(10): 6130-40, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25572395

ABSTRACT

Recent studies using ClipR-59 knock-out mice implicated this protein in the regulation of muscle function. In this report, we have examined the role of ClipR-59 in muscle differentiation and found that ClipR-59 knockdown in C2C12 cells suppressed myoblast fusion. To elucidate the molecular mechanism whereby ClipR-59 regulates myoblast fusion, we carried out a yeast two-hybrid screen using ClipR-59 as the bait and identified Elmo2, a member of the Engulfment and cell motility protein family, as a novel ClipR-59-associated protein. We showed that the interaction between ClipR-59 and Elmo2 was mediated by the atypical PH domain of Elmo2 and the Glu-Pro-rich domain of ClipR-59 and regulated by Rho-GTPase. We have examined the impact of ClipR-59 on Elmo2 downstream signaling and found that interaction of ClipR-59 with Elmo2 enhanced Rac1 activation. Collectively, our studies demonstrate that formation of an Elmo2·ClipR-59 complex plays an important role in myoblast fusion.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Muscle Development/genetics , Myoblasts/cytology , Animals , Cell Differentiation , Humans , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Myoblasts/metabolism , Neuropeptides/metabolism , Protein Structure, Tertiary/genetics , Signal Transduction , rac1 GTP-Binding Protein/metabolism
10.
Mol Cell ; 30(3): 260-1, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18471970

ABSTRACT

In this issue of Molecular Cell, Basak et al. (2008) identify Prl-3 as a p53-inducible gene acting in DNA damage-induced cell-cycle arrest. Both deletion and overexpression of Prl-3 can be cytostatic, adding to a growing list of genes whose expression must be carefully titrated for proper cell proliferation.


Subject(s)
Cell Proliferation , Gene Expression Regulation , Neoplasm Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Cycle/physiology , DNA Damage , Humans , Neoplasm Proteins/genetics , Protein Tyrosine Phosphatases/genetics , Tumor Suppressor Protein p53/genetics
11.
J Biol Chem ; 289(18): 12520-34, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24659782

ABSTRACT

Angiogenin (ANG) and ribonuclease 4 (RNASE4), two members of the secreted and vertebrate-specific ribonuclease superfamily, play important roles in cancers and neurodegenerative diseases. The ANG and RNASE4 genes share genetic regions with promoter activities, but the structure and regulation of these putative promotes are unknown. We have characterized the promoter regions, defined the transcription start site, and identified a mechanism of transcription regulation that involves both RNA polymerase III (Pol III) elements and CCCTC binding factor (CTCF) sites. We found that two Pol III elements within the promoter region influence ANG and RNASE4 expression in a position- and orientation-dependent manner. We also provide evidence for the presence of an intragenic chromatin loop between the two CTCF binding sites located in two introns flanking the ANG coding exon. We found that formation of this intragenic loop preferentially enhances ANG transcription. These results suggest a multilayer transcriptional regulation of ANG and RNASE4 gene locus. These data also add more direct evidence to the notion that Pol III elements are able to directly influence Pol II gene transcription. Furthermore, our data indicate that a CTCF-dependent chromatin loop is able to differentially regulate transcription of genes that share the same promoters.


Subject(s)
Chromatin/genetics , RNA Polymerase III/metabolism , Repressor Proteins/metabolism , Ribonuclease, Pancreatic/genetics , Ribonucleases/genetics , Transcription, Genetic , Binding Sites/genetics , CCCTC-Binding Factor , Cell Line, Tumor , Chromatin/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Mutation , Promoter Regions, Genetic/genetics , RNA Interference , RNA Polymerase II/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism , Transcription Initiation Site
12.
Cancer Cell ; 9(1): 13-22, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16413468

ABSTRACT

Cyclin D1 is a multifunctional protein that activates CDK4 and CDK6, titrates Cip/Kip CDK inhibitors to increase CDK2 activity, and modulates the function of certain transcription factors. To specifically test the importance of cyclin D1-associated kinase activity, we generated "knockin" mice expressing mutant cyclin D1 deficient in activating CDK4/6. The development of several cyclin D1-dependent compartments, including mammary glands, proceeds relatively normally in these animals, demonstrating that cyclin D1-associated kinase activity is largely dispensable for development of these tissues. Strikingly, knockin mice were resistant to breast cancers initiated by ErbB-2. These results demonstrate a differential requirement for cyclin D1-CDK4/6 kinase activity in development versus tumorigenesis and strongly support cyclin D1-dependent kinase activity as a specific therapeutic target in breast cancer.


Subject(s)
Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Mammary Glands, Animal/growth & development , Mammary Neoplasms, Experimental/metabolism , Animals , Cyclin D1/genetics , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/genetics , Enzyme Activation , Female , Genes, erbB-2 , Mammary Glands, Animal/enzymology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Mutant Strains , Mutation , Protein Binding , Retina/enzymology , Retina/growth & development
13.
J Biol Chem ; 287(29): 24330-8, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22573325

ABSTRACT

Matrix metalloprotease-1 (MMP1), a collagenase and activator of the G protein-coupled protease activated receptor-1 (PAR1), is an emerging new target implicated in oncogenesis and metastasis in diverse cancers. However, the functional mouse homologue of MMP1 in cancer models has not yet been clearly defined. We report here that Mmp1a is a functional MMP1 homologue that promotes invasion and metastatic progression of mouse lung cancer and melanoma. LLC1 (Lewis lung carcinoma) and primary mouse melanoma cells harboring active BRAF express high levels of endogenous Mmp1a, which is required for invasion through collagen. Silencing of either Mmp1a or PAR1 suppressed invasive stellate growth of lung cancer cells in three-dimensional matrices. Conversely, ectopic expression of Mmp1a conferred an invasive phenotype in epithelial cells that do not express endogenous Mmp1a. Consistent with Mmp1a acting as a PAR1 agonist in an autocrine loop, inhibition or silencing of PAR1 resulted in a loss of the Mmp1a-driven invasive phenotype. Knockdown of Mmp1a on tumor cells resulted in significantly decreased tumorigenesis, invasion, and metastasis in xenograft models. Together, these data demonstrate that cancer cell-derived Mmp1a acts as a robust functional homologue of MMP1 by conferring protumorigenic and metastatic behavior to cells.


Subject(s)
Matrix Metalloproteinase 1/metabolism , Animals , Cell Line , Cell Line, Tumor , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Matrix Metalloproteinase 1/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Receptor, PAR-1/metabolism , Xenograft Model Antitumor Assays
14.
Blood ; 117(23): 6120-31, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21508411

ABSTRACT

Cyclin-dependent kinase-6 (CDK6) is required for early thymocyte development and tumorigenesis. To mechanistically dissect the role of CDK6 in thymocyte development, we generated and analyzed mutant knock-in mice and found that mice expressing a kinase-dead Cdk6 allele (Cdk6(K43M)) had a pronounced reduction in thymocytes and hematopoietic stem cells and progenitor cells (Lin⁻Sca-1⁺c-Kit⁺ [LSK]). In contrast, mice expressing the INK4-insensitive, hyperactive Cdk6(R31C) allele displayed excess proliferation in LSK and thymocytes. However, this is countered at least in part by increased apoptosis, which may limit progenitor and thymocyte expansion in the absence of other genetic events. Our mechanistic studies demonstrate that CDK6 kinase activity contributes to Notch signaling because inactive CDK6 kinase disrupts Notch-dependent survival, proliferation, and differentiation of LSK, with concomitant alteration of Notch target gene expression, such as massive up-regulation of CD25. Further, knockout of CD25 in Cdk6(K43M) mice rescued most defects observed in young mice. These results illustrate an important role for CDK6 kinase activity in thymocyte development that operates partially through modulating Notch target gene expression. This role of CDK6 as a downstream mediator of Notch identifies CDK6 kinase activity as a potential therapeutic target in human lymphoid malignancies.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation , Cyclin-Dependent Kinase 6/biosynthesis , Gene Expression Regulation, Enzymologic/physiology , Signal Transduction/physiology , Thymus Gland/enzymology , Alleles , Animals , Cell Survival/physiology , Cyclin-Dependent Kinase 6/genetics , Gene Knock-In Techniques , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/therapy , Humans , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/genetics , Mice , Mice, Knockout , Receptors, Notch/genetics , Receptors, Notch/metabolism
15.
Cancer Cell ; 7(6): 501-2, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15950898

ABSTRACT

The retinoblastoma protein or its regulators are altered in most human cancers. Although commonly thought of as solely a repressor of E2F-dependent transcription and cell cycle progression, pRb has gained notoriety in recent years as a key actor in cellular differentiation programs. In the June issue of Molecular Cell, Benevolenskaya et al. report that a long-known but poorly understood pRb interactor, RBP2, acts as an inhibitor of differentiation contributing to pRb's role as a coordinator of differentiation and cell cycle exit. Loss of pRb may unleash RBP2, maintaining cells in a poorly differentiated progenitor state that is prerequisite to tumor formation.


Subject(s)
Cell Differentiation/physiology , Intracellular Signaling Peptides and Proteins/physiology , Neoplasms/physiopathology , Retinoblastoma Protein/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Cycle Proteins/physiology , Cell Proliferation , DNA-Binding Proteins/physiology , E2F Transcription Factors , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Models, Biological , Neoplasms/etiology , Neoplasms/metabolism , Protein Binding , Retinoblastoma Protein/metabolism , Retinoblastoma-Binding Protein 2 , Transcription Factors/physiology , Tumor Suppressor Proteins/metabolism
16.
bioRxiv ; 2023 Aug 24.
Article in English | MEDLINE | ID: mdl-37662310

ABSTRACT

Despite recent advances in treatment, melanoma remains the deadliest form of skin cancer, due to its highly metastatic nature. Melanomas harboring oncogenic BRAF V600E mutations combined with PTEN loss exhibit unrestrained PI3K/AKT signaling and increased invasiveness. However, the contribution of different AKT isoforms to melanoma initiation, progression, and metastasis has not been comprehensively explored, and questions remain whether individual isoforms play distinct or redundant roles in each step. We investigate the contribution of individual AKT isoforms to melanoma initiation using a novel mouse model of AKT isoform-specific loss in a murine melanoma model, and investigate tumor progression, maintenance, and metastasis among a panel of human metastatic melanoma cell lines using AKT-isoform specific knockdown studies. We elucidate that AKT2 is dispensable for primary tumor formation but promotes migration and invasion in vitro and metastatic seeding in vivo , while AKT1 is uniquely important for melanoma initiation and cell proliferation. We propose a mechanism whereby inhibition of AKT2 impairs glycolysis and reduces an EMT-related gene expression signature in PTEN-null BRAF-mutant human melanoma cells to limit metastatic spread. Our data suggest that elucidation of AKT2-specific functions in metastasis could inform therapeutic strategies to improve treatment options for melanoma patients.

17.
Cancers (Basel) ; 15(20)2023 Oct 12.
Article in English | MEDLINE | ID: mdl-37894325

ABSTRACT

Despite recent advances in treatment, melanoma remains the deadliest form of skin cancer due to its highly metastatic nature. Melanomas harboring oncogenic BRAFV600E mutations combined with PTEN loss exhibit unrestrained PI3K/AKT signaling and increased invasiveness. However, the contribution of different AKT isoforms to melanoma initiation, progression, and metastasis has not been comprehensively explored, and questions remain about whether individual isoforms play distinct or redundant roles in each step. We investigate the contribution of individual AKT isoforms to melanoma initiation using a novel mouse model of AKT isoform-specific loss in a murine melanoma model, and we investigate tumor progression, maintenance, and metastasis among a panel of human metastatic melanoma cell lines using AKT isoform-specific knockdown studies. We elucidate that AKT2 is dispensable for primary tumor formation but promotes migration and invasion in vitro and metastatic seeding in vivo, whereas AKT1 is uniquely important for melanoma initiation and cell proliferation. We propose a mechanism whereby the inhibition of AKT2 impairs glycolysis and reduces an EMT-related gene expression signature in PTEN-null BRAF-mutant human melanoma cells to limit metastatic spread. Our data suggest that the elucidation of AKT2-specific functions in metastasis might inform therapeutic strategies to improve treatment options for melanoma patients.

18.
Dev Cell ; 12(4): 477-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17419986

ABSTRACT

The spindle checkpoint ensures the proper partition of the chromosomal content of dividing cells, by controlling the transition from metaphase to anaphase. In a recent issue of Cancer Cell, Vecchione and coworkers report that the protein product of the tumor suppressor gene Lzts1 (Leucine zipper tumor suppressor-1) binds the Cdk1 phosphatase Cdc25C and stabilizes it by protecting it from proteasomal degradation (Vecchione et al., 2007). Partial or complete loss of Lzts1 downregulates Cdc25C and inhibits Cdk1 activity during mitosis, leading to premature transition from metaphase to anaphase.


Subject(s)
Aneuploidy , CDC2 Protein Kinase/metabolism , Cell Cycle , Gene Expression Regulation, Enzymologic , CDC2 Protein Kinase/genetics , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Humans , Models, Biological , Tumor Suppressor Proteins/metabolism , cdc25 Phosphatases/metabolism
19.
Cancer Cell ; 3(4): 305-7, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12726855

ABSTRACT

The prevailing view of cdk2 as a critical regulator of cell cycle progression and optimal therapeutic target in cancer cells is now challenged by the observation that tumor cells deficient in cdk2 protein and kinase activity are not impaired in proliferation.


Subject(s)
CDC2-CDC28 Kinases , Cyclin-Dependent Kinases/physiology , Protein Serine-Threonine Kinases/physiology , S Phase/physiology , Animals , Cell Division , Cyclin E/genetics , Cyclin-Dependent Kinase 2 , Gene Silencing , Humans , Neoplasms/genetics , Retinoblastoma Protein/genetics , Signal Transduction
20.
Cancers (Basel) ; 14(3)2022 Jan 23.
Article in English | MEDLINE | ID: mdl-35158840

ABSTRACT

Cellular senescence is a carefully regulated process of proliferative arrest accompanied by functional and morphologic changes. Senescence allows damaged cells to avoid neoplastic proliferation; however, the induction of the senescence-associated secretory phenotype (SASP) can promote tumor growth. The complexity of senescence may limit the efficacy of anti-neoplastic agents, such as CDK4/6 inhibitors (Cdk4/6i), that induce a senescence-like state in tumor cells. The AKT kinase family, which contains three isoforms that play both unique and redundant roles in cancer progression, is commonly hyperactive in many cancers including melanoma and has been implicated in the regulation of senescence. To interrogate the role of AKT isoforms in Cdk4/6i-induced cellular senescence, we generated isoform-specific AKT knockout human melanoma cell lines. We found that the CDK4/6i Palbociclib induced a form of senescence in these cells that was dependent on AKT1. We then evaluated the activity of the cGAS-STING pathway, recently implicated in cellular senescence, finding that cGAS-STING function was dependent on AKT1, and pharmacologic inhibition of cGAS had little effect on senescence. However, we found SASP factors to require NF-κB function, in part dependent on a stimulatory phosphorylation of IKKα by AKT1. In summary, we provide the first evidence of a novel, isoform-specific role for AKT1 in therapy-induced senescence in human melanoma cells acting through NF-κB but independent of cGAS.

SELECTION OF CITATIONS
SEARCH DETAIL