Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
Heliyon ; 8(1): e08666, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35028452

ABSTRACT

Estrogen is thought to cause proliferation of all estrogen receptor positive (ER+) breast cancers. Paradoxically, in the Women's Health Initiative Trial, estrogen-only hormone replacement therapy reduced the incidence and mortality of low grade, ER+, HER2- breast cancer. We gave estradiol to 19 post-menopausal women with newly diagnosed low-grade, ER+, HER2- breast cancer in a prospective window of opportunity clinical trial and examined the changes in proliferation and gene expression before and after estradiol treatment. Ki67 decreased in 13/19 (68%) patients and 8/13 (62%) showed a decrease in Risk of Recurrence Score. We chose three prototypical estrogen responders (greatest decrease in ROR) and non-responders (no/minimal change in ROR) and applied a differential gene expression analysis to develop pre-treatment (PRESTO-30core) and post-treatment (PRESTO-45surg) gene expression profiles. The PRESTO-30core predicted adjuvant benefit in a published series of tamoxifen, the partial estrogen agonist. Of the 45 genes in the PRESTO-45surg, thirty contain the Cell cycle genes Homology Region (CHR) motif that binds the class B multi-vulva complex (MuvB) a member of the DREAM (Dimerization partner, retinoblastoma-like proteins, E2F, MuvB) complex responsible for reversible cell cycle arrest or quiescence. There was also near uniform suppression (89%) of the remaining DREAM genes consistent with estrogen induced activation of the DREAM complex to mediate cell cycle block after a short course of estrogens. To our knowledge, this is the first report to show estrogen modulation of DREAM genes and suggest involvement of DREAM pathway associated quiescence in endocrine responsive post-menopausal ER+ breast cancers.

4.
Cancer Res ; 79(7): 1646-1657, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30659022

ABSTRACT

The mechanisms by which breast cancers progress from relatively indolent ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC) are not well understood. However, this process is critical to the acquisition of metastatic potential. MAPK-interacting serine/threonine-protein kinase 1 (MNK1) signaling can promote cell invasion. NODAL, a morphogen essential for embryogenic patterning, is often reexpressed in breast cancer. Here we describe a MNK1/NODAL signaling axis that promotes DCIS progression to IDC. We generated MNK1 knockout (KO) or constitutively active MNK1 (caMNK1)-expressing human MCF-10A-derived DCIS cell lines, which were orthotopically injected into the mammary glands of mice. Loss of MNK1 repressed NODAL expression, inhibited DCIS to IDC conversion, and decreased tumor relapse and metastasis. Conversely, caMNK1 induced NODAL expression and promoted IDC. The MNK1/NODAL axis promoted cancer stem cell properties and invasion in vitro. The MNK1/2 inhibitor SEL201 blocked DCIS progression to invasive disease in vivo. In clinical samples, IDC and DCIS with microinvasion expressed higher levels of phospho-MNK1 and NODAL versus low-grade (invasion-free) DCIS. Cumulatively, our data support further development of MNK1 inhibitors as therapeutics for preventing invasive disease. SIGNIFICANCE: These findings provide new mechanistic insight into progression of ductal carcinoma and support clinical application of MNK1 inhibitors to delay progression of indolent ductal carcinoma in situ to invasive ductal carcinoma.


Subject(s)
Breast Carcinoma In Situ/pathology , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Nodal Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Breast Carcinoma In Situ/metabolism , Breast Neoplasms/metabolism , CRISPR-Cas Systems , Carcinoma, Ductal, Breast/metabolism , Cell Line, Tumor , Cell Proliferation , Disease Progression , Female , Heterografts , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Nude , Protein Serine-Threonine Kinases/genetics
5.
J Gynecol Oncol ; 28(6): e82, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29027400

ABSTRACT

OBJECTIVE: The use of robotic radical hysterectomy has greatly increased in the treatment of early stage cervical cancer. We sought to compare surgical and oncologic outcomes of women undergoing robotic radical hysterectomy compared to open radical hysterectomy. METHODS: The clinic-pathologic, treatment, and recurrence data were abstracted through an Institutional Review Board-approved protocol at 2 separate large tertiary care centers in Seattle, Swedish Medical Center and the University of Washington. Data were collected from 2001-2012. Comparisons between the robotic and open cohorts were made for complications, recurrence, progression-free survival (PFS), and overall survival (OS). RESULTS: In the study period, 109 robotic radical hysterectomies were performed. These were compared to 202 open radical hysterectomies. The groups were comparable in terms of age and body mass index (BMI). Length of stay (LOS) was considerably shorter in the robotic group (42.7 vs. 112.6 hours, p<0.001) as was estimated blood loss (EBL; 105.9 vs. 482.6 mL, p<0.001). There were more complications in the open radical hysterectomy group, 23.4% vs. 9.2% in the robotic group (p=0.002). The recurrence rate was comparable between the groups (10.1% vs. 10.4%, p=0.730). In multivariate adjusted analysis, robotic surgery was not a statistically significant predictor of PFS (p=0.230) or OS (0.85). CONCLUSION: Our study, one of the largest multi-institution cohorts of patients undergoing robotic radical hysterectomy, suggest robotic radical hysterectomy leads to comparable oncologic outcomes in the treatment of early stage cervical cancer with improved short-term surgical outcomes such as decreased LOS and EBL.


Subject(s)
Adenocarcinoma/surgery , Carcinoma, Adenosquamous/surgery , Carcinoma, Squamous Cell/surgery , Hysterectomy/methods , Laparoscopy/methods , Neoplasm Recurrence, Local/epidemiology , Postoperative Complications/epidemiology , Robotic Surgical Procedures/methods , Uterine Cervical Neoplasms/surgery , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Blood Transfusion/statistics & numerical data , Carcinoma, Adenosquamous/pathology , Carcinoma, Squamous Cell/pathology , Disease-Free Survival , Female , Humans , Laparotomy/methods , Length of Stay , Middle Aged , Neoplasm Staging , Postoperative Complications/therapy , Survival Rate , Treatment Outcome , Tumor Burden , Uterine Cervical Neoplasms/pathology , Young Adult
6.
Clin Imaging ; 40(5): 1047-54, 2016.
Article in English | MEDLINE | ID: mdl-27379441

ABSTRACT

OBJECTIVE: To determine the underlying histopathologic features influencing apparent diffusion coefficient (ADC) values of breast fibroadenomas. MATERIALS AND METHODS: Biopsy-proven fibroadenomas (n=26) initially identified as suspicious on breast magnetic resonance imaging (MRI) were retrospectively evaluated. Histopathologic assessments of lesion cellularity and stromal type were compared with ADC measures on diffusion-weighted MRI. RESULTS: Presence of epithelial hyperplasia (increased cellularity) and dense collagenous stroma were both significantly associated with lower lesion ADC values (P=.02 and .004, respectively). CONCLUSION: Variations in epithelial cellularity and stromal type influence breast lesion ADC values and may explain the wide range of ADC measures observed in benign fibroadenomas.


Subject(s)
Breast Density , Breast Neoplasms/diagnostic imaging , Breast/diagnostic imaging , Diffusion Magnetic Resonance Imaging , Fibroadenoma/diagnostic imaging , Adult , Aged , Biopsy , Breast/pathology , Breast Neoplasms/pathology , False Positive Reactions , Female , Fibroadenoma/pathology , Humans , Middle Aged , Retrospective Studies
7.
J Virol ; 83(4): 1602-10, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19073728

ABSTRACT

Interferon (IFN)-stimulated gene 15 (ISG15) is a ubiquitin-like molecule that conjugates to target proteins via a C-terminal LRLRGG motif and has antiviral function in vivo. We used structural modeling to predict human ISG15 (hISG15) residues important for interacting with its E1 enzyme, UbE1L. Kinetic analysis revealed that mutation of arginine 153 to alanine (R153A) ablated hISG15-hUbE1L binding and transthiolation of UbcH8. Mutation of other predicted UbE1L-interacting residues had minimal effects on the transfer of ISG15 from UbE1L to UbcH8. The capacity of hISG15 R153A to form protein conjugates in 293T cells was markedly diminished. Mutation of the homologous residue in mouse ISG15 (mISG15), arginine 151, to alanine (R151A) also attenuated protein ISGylation following transfection into 293T cells. We assessed the role of ISG15-UbE1L interactions in control of virus infection by constructing double subgenomic Sindbis viruses that expressed the mISG15 R151A mutant. While expression of mISG15 protected alpha/beta-IFN-receptor-deficient (IFN-alphabetaR(-/-)) mice from lethality following Sindbis virus infection, expression of mISG15 R151A conferred no survival benefit. The R151A mutation also attenuated ISG15's ability to decrease Sindbis virus replication in IFN-alphabetaR(-/-) mice or prolong survival of ISG15(-/-) mice. The importance of UbE1L was confirmed by demonstrating that mice lacking this ISG15 E1 enzyme were highly susceptible to Sindbis virus infection. Together, these data support a role for protein conjugation in the antiviral effects of ISG15.


Subject(s)
Alphavirus Infections/immunology , Cytokines/metabolism , Protein Interaction Mapping , Sindbis Virus/immunology , Ubiquitin-Activating Enzymes/metabolism , Ubiquitins/metabolism , Amino Acid Substitution/genetics , Animals , Cell Line , Cytokines/genetics , Humans , Male , Mice , Mutagenesis, Site-Directed , Mutation, Missense , Protein Binding , Survival Analysis , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitins/genetics
8.
Cell Host Microbe ; 2(6): 404-16, 2007 Dec 13.
Article in English | MEDLINE | ID: mdl-18078692

ABSTRACT

Ubiquitin (Ub) and interferon-stimulated gene product 15 (ISG15) reversibly conjugate to proteins and mediate important innate antiviral responses. The ovarian tumor (OTU) domain represents a superfamily of predicted proteases found in eukaryotic, bacterial, and viral proteins, some of which have Ub-deconjugating activity. We show that the OTU domain-containing proteases from nairoviruses and arteriviruses, two unrelated groups of RNA viruses, hydrolyze Ub and ISG15 from cellular target proteins. This broad activity contrasts with the target specificity of known mammalian OTU domain-containing proteins. Expression of a viral OTU domain-containing protein antagonizes the antiviral effects of ISG15 and enhances susceptibility to Sindbis virus infection in vivo. We also show that viral OTU domain-containing proteases inhibit NF-kappaB-dependent signaling. Thus, the deconjugating activity of viral OTU proteases represents a unique viral strategy to inhibit Ub- and ISG15-dependent antiviral pathways.


Subject(s)
Cytokines/immunology , Immunity, Innate , Peptide Hydrolases/physiology , Protein Structure, Tertiary/physiology , Ubiquitin/immunology , Ubiquitins/immunology , Viral Proteins/physiology , Alphavirus Infections/immunology , Alphavirus Infections/virology , Amino Acid Sequence , Animals , Arterivirus/enzymology , Arterivirus/genetics , Cytokines/metabolism , Humans , Hydrolysis , Mice , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , NF-kappa B/metabolism , Nairovirus/enzymology , Nairovirus/genetics , Neoplasm Proteins/physiology , Peptide Hydrolases/chemistry , Sequence Alignment , Signal Transduction , Sindbis Virus/enzymology , Ubiquitin/metabolism , Ubiquitins/metabolism , Viral Proteins/chemistry
9.
Proc Natl Acad Sci U S A ; 104(4): 1371-6, 2007 Jan 23.
Article in English | MEDLINE | ID: mdl-17227866

ABSTRACT

Type I interferons (IFNs) play an essential role in the host response to viral infection through the induction of numerous IFN-stimulated genes (ISGs), including important antiviral molecules such as PKR, RNase L, Mx, and iNOS. Yet, additional antiviral ISGs likely exist. IFN-stimulated gene 15 (ISG15) is a ubiquitin homolog that is rapidly up-regulated after viral infection, and it conjugates to a wide array of host proteins. Although it has been hypothesized that ISG15 functions as an antiviral molecule, the initial evaluation of ISG15-deficient mice revealed no defects in their responses to vesicular stomatitis virus or lymphocytic choriomeningitis virus, leaving open the important question of whether ISG15 is an antiviral molecule in vivo. Here we demonstrate that ISG15 is critical for the host response to viral infection. ISG15-/- mice are more susceptible to influenza A/WSN/33 and influenza B/Lee/40 virus infections. ISG15-/- mice also exhibited increased susceptibility to both herpes simplex virus type 1 and murine gammaherpesvirus 68 infection and to Sindbis virus infection. The increased susceptibility of ISG15-/- mice to Sindbis virus infection was rescued by expressing wild-type ISG15, but not a mutant form of ISG15 that cannot form conjugates, from the Sindbis virus genome. The demonstration of ISG15 as a novel antiviral molecule with activity against both RNA and DNA viruses provides a target for the development of therapies against important human pathogens.


Subject(s)
Cytokines/physiology , Herpesviridae/physiology , Orthomyxoviridae/physiology , Sindbis Virus/physiology , Animals , Cytokines/genetics , Disease Susceptibility , Male , Mice , Mice, Knockout , Ubiquitins/genetics , Ubiquitins/physiology
10.
J Virol ; 79(22): 13974-83, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16254333

ABSTRACT

The innate immune response, and in particular the alpha/beta interferon (IFN-alpha/beta) system, plays a critical role in the control of viral infections. Interferons alpha and beta exert their antiviral effects through the induction of hundreds of interferon-induced (or -stimulated) genes (ISGs). While several of these ISGs have characterized antiviral functions, their actions alone do not explain all of the effects mediated by IFN-alpha/beta. To identify additional IFN-induced antiviral molecules, we utilized a recombinant chimeric Sindbis virus to express selected ISGs in IFN-alpha/beta receptor (IFN-alpha/betaR)(-/-) mice and looked for attenuation of Sindbis virus infection. Using this approach, we identified a ubiquitin homolog, interferon-stimulated gene 15 (ISG15), as having antiviral activity. ISG15 expression protected against Sindbis virus-induced lethality and decreased Sindbis virus replication in multiple organs without inhibiting the spread of virus throughout the host. We establish that, much like ubiquitin, ISG15 requires its C-terminal LRLRGG motif to form intracellular conjugates. Finally, we demonstrate that ISG15's LRLRGG motif is also required for its antiviral activity. We conclude that ISG15 can be directly antiviral.


Subject(s)
Alphavirus Infections/immunology , Cytokines/genetics , Interferon Type I/physiology , Sindbis Virus/genetics , Amino Acid Sequence , Animals , Cytokines/physiology , Disease Models, Animal , Gene Expression Regulation, Viral/immunology , Interferon Type I/deficiency , Interferon Type I/genetics , Interferon-alpha/deficiency , Interferon-alpha/genetics , Interferon-alpha/physiology , Interferon-beta/deficiency , Interferon-beta/genetics , Interferon-beta/physiology , Mice , Mice, Knockout , Sindbis Virus/pathogenicity , Transfection , Ubiquitins/genetics , Ubiquitins/physiology
11.
Biochem Biophys Res Commun ; 336(2): 496-506, 2005 Oct 21.
Article in English | MEDLINE | ID: mdl-16139798

ABSTRACT

Though the interferon-inducible protein ISG15 was one of the first ubiquitin-like modifiers to be discovered, much remains unknown about the identity of proteins conjugated to ISG15 or the biologic consequences of modification. To gain a better understanding of the cellular pathways affected by ISG15, we identified proteins targeted for ISGylation using a proteomic approach. Mass spectrometric analysis identified 76 candidate ISGylation targets in anti-ISG15 immunoprecipitates from interferon-treated mouse or human cells. Twenty-one proteins were found in both mouse and human samples, including STAT1, a known target of ISGylation. Candidates identified in both species were tested for ISGylation in a transfection system: 18 of 19 proteins tested were ISGylated in this system. Two candidates, EF-2 and VCP, were also shown to be ISGylated in an interferon-dependent manner in the absence of exogenous over-expression. Seven proteins identified from a single species, but functionally related to candidates found in both species, were also ISGylated in the over-expression system. Proteins that can be ISGylated play important roles in translation, glycolysis, stress responses, and cell motility. These data indicate that ISGylation targets proteins found in several fundamentally important cellular pathways and will contribute to understanding the physiologic role of interferon-induced ISG15 and ISG15 conjugation.


Subject(s)
Cytokines/metabolism , Fibroblasts/metabolism , Gene Expression Profiling/methods , Interferon-beta/pharmacology , Mass Spectrometry/methods , Peptide Mapping/methods , Protein Interaction Mapping/methods , Proteome/metabolism , Animals , Cells, Cultured , Fibroblasts/drug effects , Humans , Mice , Mice, Inbred C57BL , Protein Binding , Proteomics/methods , Species Specificity , U937 Cells , Ubiquitins/metabolism
12.
Mol Cell Biol ; 24(21): 9592-600, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15485925

ABSTRACT

Protein ISGylation is unique among ubiquitin-like conjugation systems in that the expression and conjugation processes are induced by specific stimuli, mainly via the alpha/beta interferon signaling pathway. It has been suggested that protein ISGylation plays a special role in the immune response, because of its interferon-signal dependency and its appearance only in higher eukaryotic organisms. Here, we report the identification of an ISG15-conjugating enzyme, Ubc8. Like other components of the protein ISGylation system (ISG15, UBE1L, and UBP43), Ubc8 is an interferon-inducible protein. Ubc8 clearly mediates protein ISGylation in transfection assays. The reduction of Ubc8 expression by small interfering RNA causes a decrease in protein ISGylation in HeLa cells upon interferon treatment. Neither UbcH7/UbcM4, the closest homologue of Ubc8 among known ubiquitin E2s, nor the small ubiquitin-like modifier E2 Ubc9 supports protein ISGylation. These findings strongly suggest that Ubc8 is a major ISG15-conjugating enzyme responsible for protein ISGylation upon interferon stimulation. Furthermore, we established an assay system to detect ISGylated target proteins by cotransfection of ISG15, UBE1L, and Ubc8 together with a target protein to be analyzed. This method provides an easy and effective way to identify new targets for the ISGylation system and will facilitate related studies.


Subject(s)
Cytokines/metabolism , Interferons/pharmacology , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitins/analogs & derivatives , Ubiquitins/metabolism , Animals , Base Sequence , Cells, Cultured , Cytokines/chemistry , Cytokines/genetics , DNA-Binding Proteins/metabolism , Humans , Mice , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Binding , Protein Processing, Post-Translational , RNA Interference , STAT1 Transcription Factor , Signal Transduction/drug effects , Trans-Activators/metabolism , Two-Hybrid System Techniques , Ubiquitin-Activating Enzymes/deficiency , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitins/chemistry , Ubiquitins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...