Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Commun Biol ; 7(1): 426, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38589567

ABSTRACT

Wilms tumor (WT) is the most common renal malignancy of childhood. Despite improvements in the overall survival, relapse occurs in ~15% of patients with favorable histology WT (FHWT). Half of these patients will succumb to their disease. Identifying novel targeted therapies remains challenging in part due to the lack of faithful preclinical in vitro models. Here we establish twelve patient-derived WT cell lines and demonstrate that these models faithfully recapitulate WT biology using genomic and transcriptomic techniques. We then perform loss-of-function screens to identify the nuclear export gene, XPO1, as a vulnerability. We find that the FDA approved XPO1 inhibitor, KPT-330, suppresses TRIP13 expression, which is required for survival. We further identify synergy between KPT-330 and doxorubicin, a chemotherapy used in high-risk FHWT. Taken together, we identify XPO1 inhibition with KPT-330 as a potential therapeutic option to treat FHWTs and in combination with doxorubicin, leads to durable remissions in vivo.


Subject(s)
Hydrazines , Kidney Neoplasms , Triazoles , Wilms Tumor , Humans , Exportin 1 Protein , Active Transport, Cell Nucleus , Karyopherins/genetics , Karyopherins/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Cell Line, Tumor , Apoptosis , Neoplasm Recurrence, Local , Doxorubicin/pharmacology , Wilms Tumor/drug therapy , Wilms Tumor/genetics , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Cell Cycle Proteins/metabolism
2.
Chem Sci ; 15(13): 4763-4769, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38550703

ABSTRACT

Aldehydes, pervasive in various environments, pose health risks at elevated levels due to their collective toxic effects via shared mechanisms. Monitoring total aldehyde content in living systems is crucial due to their cumulative impact. Current methods for detecting cellular aldehydes are limited to UV and visible ranges, restricting their analysis in living systems. This study introduces an innovative reaction-based trigger that leverages the exceptional selectivity of 2-aminothiophenol for aldehydes, leading to the production of dihydrobenzothiazole and activating a fluorescence response. Using this trigger, we developed a series of fluorescent probes for aldehydes by altering the fluorophore allowing for excitation and emission wavelengths across the visible to near-infrared spectral regions without compromising the reactivity of the bioorthogonal moiety. These probes exhibit remarkable aldehyde chemoselectivity, rapid kinetics, and high quantum yields, enabling the detection of diverse aldehyde types, both exogenous and endogenous, within complex biological contexts. Notably, we employed the most red-shifted near-infrared probe from this series to detect aldehydes in living systems, including biliary organoids and mouse organs. These probes provide valuable tools for exploring the multifaceted roles of aldehydes in biological functions and diseases within living systems, laying the groundwork for further investigations.

3.
J Fungi (Basel) ; 9(12)2023 Dec 11.
Article in English | MEDLINE | ID: mdl-38132788

ABSTRACT

Understanding the molecular basis of cancer initiation and progression is critical in developing effective treatment strategies. Recently, mutations in genes encoding histone proteins that drive oncogenesis have been identified, converting these essential proteins into "oncohistones". Understanding how oncohistone mutants, which are commonly single missense mutations, subvert the normal function of histones to drive oncogenesis requires defining the functional consequences of such changes. Histones genes are present in multiple copies in the human genome with 15 genes encoding histone H3 isoforms, the histone for which the majority of oncohistone variants have been analyzed thus far. With so many wildtype histone proteins being expressed simultaneously within the oncohistone, it can be difficult to decipher the precise mechanistic consequences of the mutant protein. In contrast to humans, budding and fission yeast contain only two or three histone H3 genes, respectively. Furthermore, yeast histones share ~90% sequence identity with human H3 protein. Its genetic simplicity and evolutionary conservation make yeast an excellent model for characterizing oncohistones. The power of genetic approaches can also be exploited in yeast models to define cellular signaling pathways that could serve as actionable therapeutic targets. In this review, we focus on the value of yeast models to serve as a discovery tool that can provide mechanistic insights and inform subsequent translational studies in humans.

4.
Nat Neurosci ; 26(8): 1339-1351, 2023 08.
Article in English | MEDLINE | ID: mdl-37460808

ABSTRACT

Extrinsic signaling between diverse cell types is crucial for nervous system development. Ligand binding is a key driver of developmental processes. Nevertheless, it remains a significant challenge to disentangle which and how extrinsic signals act cooperatively to affect changes in recipient cells. In the developing human brain, cortical progenitors transition from neurogenesis to gliogenesis in a stereotyped sequence that is in part influenced by extrinsic ligands. Here we used published transcriptomic data to identify and functionally test five ligand-receptor pairs that synergistically drive human astrogenesis. We validate the synergistic contributions of TGFß2, NLGN1, TSLP, DKK1 and BMP4 ligands on astrocyte development in both hCOs and primary fetal tissue. We confirm that the cooperative capabilities of these five ligands are greater than their individual capacities. Additionally, we discovered that their combinatorial effects converge in part on the mTORC1 signaling pathway, resulting in transcriptomic and morphological features of astrocyte development. Our data-driven framework can leverage single-cell and bulk genomic data to generate and test functional hypotheses surrounding cell-cell communication regulating neurodevelopmental processes.


Subject(s)
Astrocytes , Neurogenesis , Humans , Astrocytes/metabolism , Ligands , Neurogenesis/physiology , Signal Transduction/physiology , Brain
5.
Front Cell Dev Biol ; 10: 909557, 2022.
Article in English | MEDLINE | ID: mdl-36060800

ABSTRACT

The world's population with obesity is reaching pandemic levels. If current trends continue, it is predicted that there will be 1.5 billion people with obesity by 2030. This projection is alarming due to the association of obesity with numerous diseases including cancer, with recent studies demonstrating a positive association with acute myeloid leukemia (AML) and B cell acute lymphoblastic leukemia (B-ALL). Interestingly, several epidemiological studies suggest the converse relationship may exist in patients with T cell acute lymphoblastic leukemia (T-ALL). To determine the relationship between obesity and T-ALL development, we employed the diet-induced obesity (DIO) murine model and cultured human T-ALL cells in adipocyte-conditioned media (ACM), bone marrow stromal cell-conditioned media, stromal conditioned media (SCM), and unconditioned media to determine the functional impact of increased adiposity on leukemia progression. Whereas only 20% of lean mice transplanted with T-ALL cells survived longer than 3 months post-inoculation, 50%-80% of obese mice with leukemia survived over this same period. Furthermore, culturing human T-ALL cells in ACM resulted in increased histone H3 acetylation (K9/K14/K18/K23/K27) and methylation (K4me3 and K27me3) posttranslational modifications (PTMs), which preceded accelerated cell cycle progression, DNA damage, and cell death. Adipocyte-mediated epigenetic changes in human T-ALL cells were recapitulated with the H3K27 demethylase inhibitor GSK-J4 and the pan-HDAC inhibitor vorinostat. These drugs were also highly cytotoxic to human T-ALL cells at low micromolar concentrations. In summary, our data support epidemiological studies demonstrating that adiposity suppresses T-ALL pathogenesis. We present data demonstrating that T-ALL cell death in adipose-rich microenvironments is induced by epigenetic modifications, which are not tolerated by leukemia cells. Similarly, GSK-J4 and vorinostat treatment induced epigenomic instability and cytotoxicity profiles that phenocopied the responses of human T-ALL cells to ACM, which provides additional support for the use of epigenetic modifying drugs as a treatment option for T-ALL.

6.
Front Oncol ; 12: 903830, 2022.
Article in English | MEDLINE | ID: mdl-35747808

ABSTRACT

Medulloblastoma (MB) is the most common malignant brain tumor in children with standard of care consisting of surgery, radiation, and chemotherapy. Recent molecular profiling led to the identification of four molecularly distinct MB subgroups - Wingless (WNT), Sonic Hedgehog (SHH), Group 3, and Group 4. Despite genomic MB characterization and subsequent tumor stratification, clinical treatment paradigms are still largely driven by histology, degree of surgical resection, and presence or absence of metastasis rather than molecular profile. Patients usually undergo resection of their tumor followed by craniospinal radiation (CSI) and a 6 month to one-year multi-agent chemotherapeutic regimen. While there is clearly a need for development of targeted agents specific to the molecular alterations of each patient, targeting proteins responsible for DNA damage repair could have a broader impact regardless of molecular subgrouping. DNA damage response (DDR) protein inhibitors have recently emerged as targeted agents with potent activity as monotherapy or in combination in different cancers. Here we discuss the molecular underpinnings of genomic instability in MB and potential avenues for exploitation through DNA damage response inhibition.

7.
G3 (Bethesda) ; 12(7)2022 07 06.
Article in English | MEDLINE | ID: mdl-35567477

ABSTRACT

Somatic missense mutations in histone genes turn these essential proteins into oncohistones, which can drive oncogenesis. Understanding how missense mutations alter histone function is challenging in mammals as mutations occur in a single histone gene. For example, described oncohistone mutations predominantly occur in the histone H3.3 gene, despite the human genome encoding 15 H3 genes. To understand how oncogenic histone missense mutations alter histone function, we leveraged the budding yeast model, which contains only 2 H3 genes, to explore the functional consequences of oncohistones H3K36M, H3G34W, H3G34L, H3G34R, and H3G34V. Analysis of cells that express each of these variants as the sole copy of H3 reveals that H3K36 mutants show different drug sensitivities compared to H3G34 mutants. This finding suggests that changes to proximal amino acids in the H3 N-terminal tail alter distinct biological pathways. We exploited the caffeine-sensitive growth of H3K36-mutant cells to perform a high copy suppressor screen. This screen identified genes linked to histone function and transcriptional regulation, including Esa1, a histone H4/H2A acetyltransferase; Tos4, a forkhead-associated domain-containing gene expression regulator; Pho92, an N6-methyladenosine RNA-binding protein; and Sgv1/Bur1, a cyclin-dependent kinase. We show that the Esa1 lysine acetyltransferase activity is critical for suppression of the caffeine-sensitive growth of H3K36R-mutant cells while the previously characterized binding interactions of Tos4 and Pho92 are not required for suppression. This screen identifies pathways that could be altered by oncohistone mutations and highlights the value of yeast genetics to identify pathways altered by such mutations.


Subject(s)
Histones , Saccharomyces cerevisiae Proteins , Animals , Caffeine , Carcinogenesis/genetics , Histone Acetyltransferases/metabolism , Histones/metabolism , Humans , Mammals , Mutation , Mutation, Missense , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
8.
Cancer Res Commun ; 2(12): 1569-1578, 2022 12.
Article in English | MEDLINE | ID: mdl-36970726

ABSTRACT

The high frequency of aberrant PI3K pathway activation in hormone receptor-positive (HR+) breast cancer has led to the development, clinical testing, and approval of the p110α-selective PI3K inhibitor alpelisib. The limited clinical efficacy of alpelisib and other PI3K inhibitors is partially attributed to the functional antagonism between PI3K and estrogen receptor (ER) signaling, which is mitigated via combined PI3K inhibition and endocrine therapy. We and others have previously demonstrated chromatin-associated mechanisms by which PI3K supports cancer development and antagonizes ER signaling through the modulation of the H3K4 methylation axis, inhibition of KDM5A promoter H3K4 demethylation and KMT2D/MLL4-directed enhancer H3K4 methylation. Here we show that inhibition of the H3K4 histone methyltransferase MLL1 in combination with PI3K inhibition impairs HR+ breast cancer clonogenicity and cell proliferation. While combined PI3K/MLL1 inhibition reduces PI3K/AKT signaling and H3K4 methylation, MLL1 inhibition increases PI3K/AKT signaling through the dysregulation of gene expression associated with AKT activation. These data reveal a feedback loop between MLL1 and AKT whereby MLL1 inhibition reactivates AKT. We show that combined PI3K and MLL1 inhibition synergizes to cause cell death in in vitro and in vivo models of HR+ breast cancer, which is enhanced by the additional genetic ablation of the H3K4 methyltransferase and AKT target KMT2D/MLL4. Together, our data provide evidence of a feedback mechanism connecting histone methylation with AKT and may support the preclinical development and testing of pan-MLL inhibitors. Significance: Here the authors leverage PI3K/AKT-driven chromatin modification to identify histone methyltransferases as a therapeutic target. Dual PI3K and MLL inhibition synergize to reduce clonogenicity and cell proliferation, and promote in vivo tumor regression. These findings suggest patients with PIK3CA-mutant, HR+ breast cancer may derive clinical benefit from combined PI3K/MLL inhibition.


Subject(s)
Breast Neoplasms , Phosphatidylinositol 3-Kinases , Humans , Female , Phosphatidylinositol 3-Kinases/genetics , Breast Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Chromatin , Histone-Lysine N-Methyltransferase/genetics , Retinoblastoma-Binding Protein 2/metabolism
9.
J Biol Chem ; 297(3): 101062, 2021 09.
Article in English | MEDLINE | ID: mdl-34375642

ABSTRACT

In 2020, the American Society of Biochemistry and Molecular Biology (ASBMB) Women in Biochemistry and Molecular Biology Committee introduced the ASBMB Leadership Awards to recognize individuals with a strong commitment to advancing the careers of women in biochemistry and molecular biology along with demonstrated excellence in research, discovery, and/or service. This innovative award recognizes efforts to mentor and support trainees and colleagues at all levels. Such a leadership award provides the opportunity to focus briefly on the important role of mentoring within the STEM disciplines. The goal of this commentary, which brings together perspectives from a senior scientist and recent recipient of the ASBMB Mid-Career Leadership Award as well as two junior faculty, is to highlight approaches for purposeful support of colleagues, with an emphasis on going beyond formal mentoring committees. The commentary primarily focuses on mentoring within the academic arena of extramural funding and publication, highlighting the reality that multiple mentors with diverse expertise and perspectives are critical to support success within STEM careers.


Subject(s)
Mentoring/methods , Mentoring/trends , Mentors/psychology , Faculty , Humans , Research Personnel , Sexism/prevention & control , Sexism/trends , United States
10.
Proc Natl Acad Sci U S A ; 117(39): 24427-24433, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32929011

ABSTRACT

PIK3CA hotspot mutation is well established as an oncogenic driver event in cancer and its durable and efficacious inhibition is a focus in the development and testing of clinical cancer therapeutics. However, hundreds of cancer-associated PIK3CA mutations remain uncharacterized, their sensitivity to PI3K inhibitors unknown. Here, we describe a series of PIK3CA C-terminal mutations, primarily nucleotide insertions, that produce a frame-shifted protein product with an extended C terminus. We report that these mutations occur at a low frequency across multiple cancer subtypes, including breast, and are sufficient to drive oncogenic transformation in vitro and in vivo. We demonstrate that the oncogenicity of these mutant p110α proteins is dependent on p85 but not Ras association. P110α-selective pharmacologic inhibition blocks transformation in cells and mammary tumors characterized by PIK3CA C-terminal mutation. Taken together, these results suggest patients with breast and other tumors characterized by PIK3CA C-terminal frameshift mutations may derive benefit from p110α-selective inhibitors, including the recently FDA-approved alpelisib.


Subject(s)
Breast Neoplasms/enzymology , Class I Phosphatidylinositol 3-Kinases/chemistry , Class I Phosphatidylinositol 3-Kinases/genetics , Frameshift Mutation , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/metabolism , Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Female , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Domains
11.
J Mol Med (Berl) ; 95(8): 791-798, 2017 08.
Article in English | MEDLINE | ID: mdl-28589435

ABSTRACT

Receptor tyrosine kinase (RTK) signaling cascades coordinate intracellular signaling in response to growth factors, chemokines, and other extracellular stimuli to control fundamental biological processes such as cellular proliferation, metabolism, and survival. Hyperactivation of pathways associated with growth factor signaling (e.g., RTK and downstream effectors including Ras, PI3K/AKT, and Raf) is a frequent event in human cancers, which uncouples ligand-mediated activation with signal transduction. While the contributions of direct genomic events are well understood as causative agents of hyperactive signal transduction, other non-heritable genomic modifications promote the activation of growth factor-associated signaling cascades. In this review, we highlight epigenomic mechanisms by which hyperactivation of RTK-associated signaling cascades occurs and may contribute to cancer.


Subject(s)
Receptor Protein-Tyrosine Kinases/genetics , Animals , Antineoplastic Agents, Hormonal/therapeutic use , DNA Methylation , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Genes, Tumor Suppressor , Histone Deacetylases/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction
12.
Proc Natl Acad Sci U S A ; 114(27): 7095-7100, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28630349

ABSTRACT

Mutation or loss of the p85 regulatory subunit of phosphatidylinositol 3-kinase (PI3K) is emerging as a transforming factor in cancer, but the mechanism of transformation has been controversial. Here we find that hemizygous deletion of the PIK3R1 gene encoding p85α is a frequent event in breast cancer, with PIK3R1 expression significantly reduced in breast tumors. PIK3R1 knockdown transforms human mammary epithelial cells, and genetic ablation of Pik3r1 accelerates a mouse model of HER2/neu-driven breast cancer. We demonstrate that partial loss of p85α increases the amount of p110α-p85 heterodimers bound to active receptors, augmenting PI3K signaling and oncogenic transformation. Pan-PI3K and p110α-selective pharmacological inhibition effectively blocks transformation driven by partial p85α loss both in vitro and in vivo. Together, our data suggest that p85α plays a tumor-suppressive role in transformation, and suggest that p110α-selective therapeutics may be effective in the treatment of breast cancer patients with PIK3R1 loss.


Subject(s)
Breast Neoplasms/metabolism , Class I Phosphatidylinositol 3-Kinases/metabolism , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Enzyme Activation , Epithelial Cells/metabolism , Female , Gene Knockdown Techniques , Genotype , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mammary Glands, Animal/metabolism , Mice , Mutation , Oligonucleotide Array Sequence Analysis , Signal Transduction
13.
Biochim Biophys Acta Rev Cancer ; 1868(1): 123-131, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28315368

ABSTRACT

The PI3-kinase/AKT pathway integrates signals from external cellular stimuli to regulate essential cellular functions, and is frequently aberrantly activated in human cancers. Recent research demonstrates that tight regulation of the epigenome is critical in preserving and restricting transcriptional activation, which can impact cellular growth and proliferation. In this review we examine mechanisms by which the PI3K/AKT pathway regulates the epigenome to promote oncogenesis, and highlight how connections between PI3K/AKT and the epigenome may impact the future therapeutic treatment of cancers featuring a hyperactivated PI3K/AKT pathway.


Subject(s)
Epigenesis, Genetic/genetics , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Animals , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Humans , Signal Transduction/genetics
14.
Cell Rep ; 15(12): 2692-704, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27292631

ABSTRACT

Post-translational histone H3 modifications regulate transcriptional competence. The mechanisms by which the epigenome is regulated in response to oncogenic signaling remain unclear. Here we show that H3K4me3 is increased in breast tumors driven by an activated PIK3CA allele and that inhibition of PI3K/AKT signaling reduces promoter-associated H3K4me3 in human breast cancer cells. We show that the H3K4 demethylase KDM5A is an AKT target and that phosphorylation of KDM5A regulates its nuclear localization and promoter occupancy. Supporting a role for KDM5A in mediating PI3K/AKT transcriptional effects, the decreased expression in response to AKT inhibition of a subset of cell-cycle genes associated with poor clinical outcome is blunted by KDM5A silencing. Our data identify a mechanism by which PI3K/AKT signaling modulates the cancer epigenome through controlling H3K4 methylation and suggest that KDM5A subcellular localization and genome occupancy may be pharmacodynamic markers of the activity of PI3K/AKT inhibitors currently in clinical development.


Subject(s)
Breast Neoplasms/metabolism , Histones/metabolism , Lysine/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Amino Acid Sequence , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle/genetics , Enzyme Activation , Female , Gene Expression Regulation, Neoplastic , Humans , Methylation , Mice , Models, Biological , Promoter Regions, Genetic , Protein Transport , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Retinoblastoma-Binding Protein 2/chemistry , Retinoblastoma-Binding Protein 2/metabolism , Subcellular Fractions/enzymology , Substrate Specificity , Treatment Outcome
15.
PLoS Pathog ; 9(3): e1003237, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23516367

ABSTRACT

The high-risk human papillomavirus (HPV) E6 proteins are consistently expressed in HPV-associated lesions and cancers. HPV16 E6 sustains the activity of the mTORC1 and mTORC2 signaling cascades under conditions of growth factor deprivation. Here we report that HPV16 E6 activated mTORC1 by enhanced signaling through receptor protein tyrosine kinases, including epidermal growth factor receptor and insulin receptor and insulin-like growth factor receptors. This is evidenced by sustained signaling through these receptors for several hours after growth factor withdrawal. HPV16 E6 increased the internalization of activated receptor species, and the signaling adaptor protein GRB2 was shown to be critical for HPV16 E6 mediated enhanced EGFR internalization and mTORC1 activation. As a consequence of receptor protein kinase mediated mTORC1 activation, HPV16 E6 expression increased cellular migration of primary human epithelial cells. This study identifies a previously unappreciated mechanism by which HPV E6 proteins perturb host-signaling pathways presumably to sustain protein synthesis during the viral life cycle that may also contribute to cellular transforming activities of high-risk HPV E6 proteins.


Subject(s)
Human papillomavirus 16/physiology , Multiprotein Complexes/metabolism , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections/metabolism , Repressor Proteins/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Uterine Cervical Neoplasms/metabolism , Cell Line , Cell Movement , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Human papillomavirus 16/genetics , Humans , Infant, Newborn , Keratinocytes/cytology , Keratinocytes/metabolism , Keratinocytes/virology , Male , Mechanistic Target of Rapamycin Complex 1 , Models, Biological , Multiprotein Complexes/genetics , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/virology , Phosphorylation , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Receptors, Somatomedin/genetics , Receptors, Somatomedin/metabolism , Repressor Proteins/genetics , TOR Serine-Threonine Kinases/genetics , Uterine Cervical Neoplasms/virology
16.
J Virol ; 87(8): 4762-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23365452

ABSTRACT

Cutaneous ß-human papillomavirus (ß-HPV) E6 proteins inhibit NOTCH signaling by associating with the transcriptional coactivator MAML1. NOTCH has tumor suppressor activities in epithelial cells and is activated during keratinocyte differentiation. Here we report that HPV type 8 (HPV8) E6 subverts NOTCH activation during keratinocyte differentiation by inhibiting RBPJ/MAML1 transcriptional activator complexes at NOTCH target DNA. NOTCH inhibition impairs epithelial differentiation and may thus contribute to ß-HPV replication and viral oncogenesis.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Keratinocytes/virology , Oncogene Proteins, Viral/metabolism , Papillomaviridae/pathogenicity , Receptors, Notch/antagonists & inhibitors , Transcription Factors/metabolism , Cells, Cultured , Humans , Keratinocytes/physiology , Virulence Factors/metabolism
17.
Nature ; 487(7408): 491-5, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22810586

ABSTRACT

Genotypic differences greatly influence susceptibility and resistance to disease. Understanding genotype-phenotype relationships requires that phenotypes be viewed as manifestations of network properties, rather than simply as the result of individual genomic variations. Genome sequencing efforts have identified numerous germline mutations, and large numbers of somatic genomic alterations, associated with a predisposition to cancer. However, it remains difficult to distinguish background, or 'passenger', cancer mutations from causal, or 'driver', mutations in these data sets. Human viruses intrinsically depend on their host cell during the course of infection and can elicit pathological phenotypes similar to those arising from mutations. Here we test the hypothesis that genomic variations and tumour viruses may cause cancer through related mechanisms, by systematically examining host interactome and transcriptome network perturbations caused by DNA tumour virus proteins. The resulting integrated viral perturbation data reflects rewiring of the host cell networks, and highlights pathways, such as Notch signalling and apoptosis, that go awry in cancer. We show that systematic analyses of host targets of viral proteins can identify cancer genes with a success rate on a par with their identification through functional genomics and large-scale cataloguing of tumour mutations. Together, these complementary approaches increase the specificity of cancer gene identification. Combining systems-level studies of pathogen-encoded gene products with genomic approaches will facilitate the prioritization of cancer-causing driver genes to advance the understanding of the genetic basis of human cancer.


Subject(s)
Genes, Neoplasm/genetics , Genome, Human/genetics , Host-Pathogen Interactions , Neoplasms/genetics , Neoplasms/metabolism , Oncogenic Viruses/pathogenicity , Viral Proteins/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Adenoviridae/pathogenicity , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions/genetics , Humans , Neoplasms/pathology , Oncogenic Viruses/genetics , Oncogenic Viruses/metabolism , Open Reading Frames/genetics , Papillomaviridae/genetics , Papillomaviridae/metabolism , Papillomaviridae/pathogenicity , Polyomavirus/genetics , Polyomavirus/metabolism , Polyomavirus/pathogenicity , Receptors, Notch/metabolism , Signal Transduction , Two-Hybrid System Techniques , Viral Proteins/genetics
18.
J Virol ; 86(14): 7466-72, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22553330

ABSTRACT

The human papillomavirus (HPV) type 16 (HPV16) E6 protein can stimulate mechanistic target of rapamycin complex 1 (mTORC1) signaling and cap-dependent translation through activation of the PDK1 and mTORC2 kinases. Here we report that HPV18 E6 also enhances cap-dependent translation. The integrity of LXXLL and PDZ protein binding domains is important for activation of cap-dependent translation by high-risk mucosal HPV E6 proteins. Consistent with this model, low-risk mucosal HPV6b and HPV11 E6 proteins, which do not contain a PDZ protein binding motif, also activate cap-dependent translation and mTORC1, albeit at a lower efficiency than high-risk HPV E6 proteins. In contrast, cutaneous HPV5 and HPV8 E6 proteins, which lack LXXLL and PDZ motif protein binding, do not enhance cap-dependent translation. Mutagenic analyses of low-risk HPV E6 proteins revealed that association with the LXXLL motif containing ubiquitin ligase E6AP (UBE3A) correlates with activation of cap-dependent translation. Hence, activation of mTORC1 and cap-dependent translation may be important for the viral life cycle in specific epithelial tissue types and contribute to cellular transformation in cooperation with other biological activities of high-risk HPV E6-containing proteins.


Subject(s)
Human papillomavirus 16/metabolism , Human papillomavirus 18/metabolism , Oncogene Proteins, Viral/metabolism , PDZ Domains , Protein Biosynthesis , Protein Interaction Domains and Motifs , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Cell Line , HEK293 Cells , Human papillomavirus 11/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Mucous Membrane/virology , Multiprotein Complexes , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , TOR Serine-Threonine Kinases , Transcription Factors/biosynthesis , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/chemistry
19.
J Virol ; 84(18): 9398-407, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20631133

ABSTRACT

The mammalian target of rapamycin (mTOR) kinase acts as a cellular rheostat that integrates signals from a variety of cellular signal transduction pathways that sense growth factor and nutrient availability as well as intracellular energy status. It was previously reported that the human papillomavirus type 16 (HPV16) E6 oncoprotein may activate the S6 protein kinase (S6K) through binding and E6AP-mediated degradation of the mTOR inhibitor tuberous sclerosis complex 2 (TSC2) (Z. Lu, X. Hu, Y. Li, L. Zheng, Y. Zhou, H. Jiang, T. Ning, Z. Basang, C. Zhang, and Y. Ke, J. Biol. Chem. 279:35664-35670, 2004; L. Zheng, H. Ding, Z. Lu, Y. Li, Y. Pan, T. Ning, and Y. Ke, Genes Cells 13:285-294, 2008). Our results confirmed that HPV16 E6 expression causes an increase in mTORC1 activity through enhanced phosphorylation of mTOR and activation of downstream signaling pathways S6K and eukaryotic initiation factor binding protein 1 (4E-BP1). However, we did not detect a decrease in TSC2 levels in HPV16 E6-expressing cells. We discovered, however, that HPV16 E6 expression causes AKT activation through the upstream kinases PDK1 and mTORC2 under conditions of nutrient deprivation. We show that HPV16 E6 expression causes an increase in protein synthesis by enhancing translation initiation complex assembly at the 5' mRNA cap and an increase in cap-dependent translation. The increase in cap-dependent translation likely results from HPV16 E6-induced AKT/mTORC1 activation, as the assembly of the translation initiation complex and cap-dependent translation are rapamycin sensitive. Lastly, coexpression of the HPV16 E6 and E7 oncoproteins does not affect HPV16 E6-induced activation of mTORC1 and cap-dependent translation. HPV16 E6-mediated activation of mTORC1 signaling and cap-dependent translation may be a mechanism to promote viral replication under conditions of limited nutrient supply in differentiated, HPV oncoprotein-expressing proliferating cells.


Subject(s)
Oncogene Proteins, Viral/metabolism , Peptide Chain Initiation, Translational , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Cell Line , Host-Pathogen Interactions , Humans , Keratinocytes/virology , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Oncogene Protein v-akt/biosynthesis , Phosphorylation , Protein Serine-Threonine Kinases/biosynthesis , Proteins , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , TOR Serine-Threonine Kinases , Transcription Factors/biosynthesis , Up-Regulation
20.
Autophagy ; 5(4): 578-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19333004

ABSTRACT

Normal human epithelial cells that express the human papillomavirus type 16 (HPV16) E7 oncoprotein show evidence of autophagy, and when deprived of growth factors they undergo cell death. We hypothesize that the process of autophagy renders such cells particularly vulnerable to cell death and suggest that autophagy induced as a consequence of uncontrolled proliferation and changes in metabolism may represent an "Achilles' heel" of human cancers and serve as a target for cancer therapy.


Subject(s)
Autophagy , Epithelial Cells/cytology , Epithelial Cells/metabolism , Neoplasms/pathology , Oncogene Proteins/metabolism , Viral Proteins/metabolism , Humans , Models, Biological , Neoplasms/virology , Neutralization Tests , Oncogene Proteins, Viral/metabolism , Papillomaviridae/metabolism , Papillomavirus E7 Proteins , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...