Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
bioRxiv ; 2023 Oct 14.
Article En | MEDLINE | ID: mdl-37873085

Mono(ADP-ribosyl)ation (MARylation), a post-translational modification (PTM) of proteins, is emerging as a critical regulator of ribosome function and translation. Herein, we demonstrate that RACK1, a member of the tryptophan-aspartate repeat (WD-repeat) family of proteins and an integral component of the ribosome, is MARylated by the mono(ADP-ribosyl) transferase (MART) PARP14 in ovarian cancer cells. We mapped and confirmed the sites of MARylation, which occur on three acidic residues within blades 4 and 5 of ß-propeller domain of RACK1, a chaperone that shuttles and anchors proteins where needed. Site-specific MARylation of RACK1 is required for stress granule formation and promotes the colocalization of RACK1 to stress granules with key components, such as G3BP1, eIF3η, and 40S ribosomal proteins. In parallel, we observed reduced translation of a subset of mRNAs, including those encoding key cancer regulators (e.g., AKT). Treatment with a PARP14 inhibitor or mutation of the sites of MARylation on RACK1 blocks these outcomes. To re-set the system after prolonged stress and recovery, the ADP-ribosyl hydrolase TARG1 deMARylates RACK1 to dissociate the stress granules and return RACK1 and the 40S ribosomal subunit to the cytoplasm, allowing for a restoration of translation. Collectively, our results highlight the discovery of a PARP14/TARG1-regulated RACK1 MARylation cycle that controls stress granule assembly and disassembly in ovarian cancer cells.

2.
Methods Mol Biol ; 2609: 75-90, 2023.
Article En | MEDLINE | ID: mdl-36515830

ADP-ribosylation (ADPRylation) is a reversible posttranslational modification resulting in the covalent attachment of ADP-ribose (ADPR) moieties on substrate proteins. Naturally occurring protein motifs and domains, including WWEs, PBZs (PAR binding zinc fingers), and macrodomains, act as "readers" for protein-linked ADPR. Although recombinant, antibody-like ADPR detection reagents containing these readers have facilitated the detection of ADPR, they are limited in their ability to capture the dynamic nature of ADPRylation. Herein, we describe the preparation and use of poly(ADP-ribose) (PAR) Trackers (PAR-Ts)-optimized dimerization-dependent or split-protein reassembly PAR sensors containing a naturally occurring PAR binding domain fused to both halves of dimerization-dependent GFP (ddGFP) or split nano luciferase (NanoLuc), respectively. We also describe how these tools can be used for the detection and quantification of PAR levels in biochemical assays with extracts and in living cells. These protocols will allow users to explore the broad utility of PAR-Ts for detecting PAR in various experimental and biological systems.


Poly Adenosine Diphosphate Ribose , Ribose , Poly Adenosine Diphosphate Ribose/metabolism , Adenosine Diphosphate Ribose/chemistry , ADP-Ribosylation , Protein Processing, Post-Translational , Recombinant Proteins/metabolism
3.
Elife ; 112022 04 27.
Article En | MEDLINE | ID: mdl-35476036

ADP-ribosylation (ADPRylation) is a reversible post-translation modification resulting in the covalent attachment of ADP-ribose (ADPR) moieties on substrate proteins. Naturally occurring protein motifs and domains, including WWEs, PBZs, and macrodomains, act as 'readers' for protein-linked ADPR. Although recombinant, antibody-like ADPR detection reagents containing these readers have facilitated the detection of ADPR, they are limited in their ability to capture the dynamic nature of ADPRylation. Herein, we describe and characterize a set of poly(ADP-ribose) (PAR) Trackers (PAR-Ts)-optimized dimerization-dependent or split-protein reassembly PAR sensors in which a naturally occurring PAR binding domain, WWE, was fused to both halves of dimerization-dependent GFP (ddGFP) or split Nano Luciferase (NanoLuc), respectively. We demonstrate that these new tools allow the detection and quantification of PAR levels in extracts, living cells, and living tissues with greater sensitivity, as well as temporal and spatial precision. Importantly, these sensors detect changes in cellular ADPR levels in response to physiological cues (e.g., hormone-dependent induction of adipogenesis without DNA damage), as well as xenograft tumor tissues in living mice. Our results indicate that PAR Trackers have broad utility for detecting ADPR in many different experimental and biological systems.


Adenosine Diphosphate Ribose , Poly Adenosine Diphosphate Ribose , ADP-Ribosylation , Adenosine Diphosphate Ribose/metabolism , Animals , DNA Damage , Humans , Mice , Poly Adenosine Diphosphate Ribose/chemistry , Poly Adenosine Diphosphate Ribose/genetics , Poly Adenosine Diphosphate Ribose/metabolism , Recombinant Proteins/metabolism
4.
Cell Chem Biol ; 29(2): 171-173, 2022 02 17.
Article En | MEDLINE | ID: mdl-35180430

In this issue of Cell Chemical Biology, Palve et al. (2022) identified PARP16 as a non-canonical therapeutic target of the PARP1 inhibitor talazoparib, which synergizes with the WEE1 inhibitor adavosertib to enhance its efficacy. The dual targeting of PARP1 and PARP16 may explain the greater efficacy of talazoparib in some cancers.


Antineoplastic Agents , Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Neoplasms/drug therapy , Phthalazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use
5.
Cell ; 184(17): 4531-4546.e26, 2021 08 19.
Article En | MEDLINE | ID: mdl-34314702

Defects in translation lead to changes in the expression of proteins that can serve as drivers of cancer formation. Here, we show that cytosolic NAD+ synthesis plays an essential role in ovarian cancer by regulating translation and maintaining protein homeostasis. Expression of NMNAT-2, a cytosolic NAD+ synthase, is highly upregulated in ovarian cancers. NMNAT-2 supports the catalytic activity of the mono(ADP-ribosyl) transferase (MART) PARP-16, which mono(ADP-ribosyl)ates (MARylates) ribosomal proteins. Depletion of NMNAT-2 or PARP-16 leads to inhibition of MARylation, increased polysome association and enhanced translation of specific mRNAs, aggregation of their translated protein products, and reduced growth of ovarian cancer cells. Furthermore, MARylation of the ribosomal proteins, such as RPL24 and RPS6, inhibits polysome assembly by stabilizing eIF6 binding to ribosomes. Collectively, our results demonstrate that ribosome MARylation promotes protein homeostasis in cancers by fine-tuning the levels of protein synthesis and preventing toxic protein aggregation.


ADP-Ribosylation , Ovarian Neoplasms/metabolism , Protein Biosynthesis , Proteostasis , Ribosomes/metabolism , 3' Untranslated Regions/genetics , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation , Endoplasmic Reticulum Stress , Fallopian Tubes/metabolism , Female , Humans , Mice, Inbred NOD , Mice, SCID , NAD/metabolism , Nicotinamide-Nucleotide Adenylyltransferase , Nucleic Acid Conformation , Ovarian Neoplasms/pathology , Poly(ADP-ribose) Polymerases/metabolism , Polyribosomes/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Ribosomal Proteins/metabolism
6.
Cells ; 10(2)2021 02 03.
Article En | MEDLINE | ID: mdl-33546365

Mono(ADP-ribosyl)ation (MARylation) is a regulatory post-translational modification of proteins that controls their functions through a variety of mechanisms. MARylation is catalyzed by mono(ADP-ribosyl) transferase (MART) enzymes, a subclass of the poly(ADP-ribosyl) polymerase (PARP) family of enzymes. Although the role of PARPs and poly(ADP-ribosyl)ation (PARylation) in cellular pathways, such as DNA repair and transcription, is well studied, the role of MARylation and MARTs (i.e., the PARP 'monoenzymes') are not well understood. Moreover, compared to PARPs, the development of MART-targeted therapeutics is in its infancy. Recent studies are beginning to shed light on the structural features, catalytic targets, and biological functions of MARTs. The development of new technologies to study MARTs have uncovered essential roles for these enzymes in the regulation of cellular processes, such as RNA metabolism, cellular transport, focal adhesion, and stress responses. These insights have increased our understanding of the biological functions of MARTs in cancers, neuronal development, and immune responses. Furthermore, several novel inhibitors of MARTs have been developed and are nearing clinical utility. In this review, we summarize the biological functions and molecular mechanisms of MARTs and MARylation, as well as recent advances in technology that have enabled detection and inhibition of their activity. We emphasize PARP-7, which is at the forefront of the MART subfamily with respect to understanding its biological roles and the development of therapeutically useful inhibitors. Collectively, the available studies reveal a growing understanding of the biochemistry, chemical biology, physiology, and pathology of MARTs.


ADP-Ribosylation/genetics , Cytosol/metabolism , Poly ADP Ribosylation/genetics , Poly Adenosine Diphosphate Ribose/metabolism , Protein Processing, Post-Translational/genetics , Humans
7.
Elife ; 102021 01 21.
Article En | MEDLINE | ID: mdl-33475085

PARP-7 (TiPARP) is a mono(ADP-ribosyl) transferase whose protein substrates and biological activities are poorly understood. We observed that PARP7 mRNA levels are lower in ovarian cancer patient samples compared to non-cancerous tissue, but PARP-7 protein nonetheless contributes to several cancer-related biological endpoints in ovarian cancer cells (e.g. growth, migration). Global gene expression analyses in ovarian cancer cells subjected to PARP-7 depletion indicate biological roles for PARP-7 in cell-cell adhesion and gene regulation. To identify the MARylated substrates of PARP-7 in ovarian cancer cells, we developed an NAD+ analog-sensitive approach, which we coupled with mass spectrometry to identify the PARP-7 ADP-ribosylated proteome in ovarian cancer cells, including cell-cell adhesion and cytoskeletal proteins. Specifically, we found that PARP-7 MARylates α-tubulin to promote microtubule instability, which may regulate ovarian cancer cell growth and motility. In sum, we identified an extensive PARP-7 ADP-ribosylated proteome with important roles in cancer-related cellular phenotypes.


Cancer is a complex illness where changes inside healthy cells causes them to grow and reproduce rapidly. Specialized proteins called enzymes ­ which regulate chemical reactions in the cell ­ often help cancer develop and spread through the body. One such enzyme called PARP-7 labels other proteins by attaching a chemical group which changes their behavior. However, it was unknown which proteins PARP-7 modifies and how this tag alters the actions of these proteins. To investigate this, Parsons, Challa, Gibson et al. developed a method to find and identify the proteins labelled by PARP-7 in ovarian cancer cells taken from patients and cultured in the laboratory. This revealed that PARP-7 labels hundreds of different proteins, including adhesion proteins which affect the connections between cells and cytoskeletal proteins which regulate a cell's shape and how it moves. One of the cytoskeletal proteins modified by PARP-7 is α-tubulin, which joins together with other tubulins to form long, tube-like structures known as microtubules. Parsons et al. found that when α-tubulin is labelled by PARP-7, it creates unstable microtubules that alter how the cancer cells grow and move. They discovered that depleting PARP-7 or mutating the sites where it modifies α-tubulin increased the stability of microtubules and slowed the growth of ovarian cancer cells. Ovarian cancer is the fifth leading cause of cancer-related deaths among women in the United States. A new drug which suppresses the activity of PARP-7 has recently been developed, and this drug could potentially be used to treat ovarian cancer patients with high levels of PARP-7. Clinical trials are ongoing to see how this drug affects the behavior of cancer cells in patients.


ADP-Ribosylation , Microtubules/metabolism , Nucleoside Transport Proteins/genetics , Cell Line, Tumor , Female , Humans , Male , Nucleoside Transport Proteins/metabolism , Ovarian Neoplasms/metabolism
8.
Cardiovasc Revasc Med ; 21(12): 1550-1554, 2020 12.
Article En | MEDLINE | ID: mdl-32546383

BACKGROUND: Intravascular brachytherapy (VBT) is an established treatment for the management of in-stent restenosis (ISR). However, whether VBT is associated with improved patient reported outcomes unknown. METHODS: We evaluated 51 consecutive patients undergoing VBT in one or more coronary arteries from January 2018 to September 2019. Data on baseline characteristics, procedural outcomes and adverse events were obtained. All patients completed the Seattle Angina Questionnaire - 7 (SAQ-7) form before and after VBT at 1 month and 6 months. RESULTS: The mean age was 69 ± 9 years and 29 (57%) of patients were males. Procedural success was 94.1%. The mean summary SAQ-7 score improved significantly (53.2 ± 21 vs. 83 ± 19, p < .001) at 30-days. The median Quality of Life (QoL) component of SAQ-7 score was 31.3 (Interquartile Range [IQR]: 18.8, 62.5) and improved to 82.5 (IQR: 62.5, 100), p < .001 at 30 days and 87.5 [IQR: 75, 100), p < .001 at last follow up. Likewise, the median angina frequency component of the SQL-7 score pre-VBT was 55 (IQR: 45, 80) and improved significantly to 90 (IQR: 60, 100) at 30-days, p < .001 and 100 [IQR: 68.8, 100], p = .02 at last follow up. Lastly, the median activity component of the SAQ-7 score improved from 83.3 (IQR: 60-100) to 100 (IQR: 83, 100), p = .01 at 30-days. Thus, results were evident as early as 1 month and sustained at median follow up of 17 months. CONCLUSION: VBT is associated with improvement in patient reported outcome measures at short term and long term follow up.


Brachytherapy , Coronary Artery Disease , Coronary Restenosis , Aged , Female , Humans , Male , Middle Aged , Patient Reported Outcome Measures , Quality of Life , Stents
9.
Genes Dev ; 34(5-6): 302-320, 2020 03 01.
Article En | MEDLINE | ID: mdl-32029452

ADP-ribosylation (ADPRylation) is a posttranslational modification of proteins discovered nearly six decades ago, but many important questions remain regarding its molecular functions and biological roles, as well as the activity of the ADP-ribose (ADPR) transferase enzymes (PARP family members) that catalyze it. Growing evidence indicates that PARP-mediated ADPRylation events are key regulators of the protein biosynthetic pathway, leading from rDNA transcription and ribosome biogenesis to mRNA synthesis, processing, and translation. In this review we describe the role of PARP proteins and ADPRylation in all facets of this pathway. PARP-1 and its enzymatic activity are key regulators of rDNA transcription, which is a critical step in ribosome biogenesis. An emerging role of PARPs in alternative splicing of mRNAs, as well as direct ADPRylation of mRNAs, highlight the role of PARP members in RNA processing. Furthermore, PARP activity, stimulated by cellular stresses, such as viral infections and ER stress, leads to the regulation of mRNA stability and protein synthesis through posttranscriptional mechanisms. Dysregulation of PARP activity in these processes can promote disease states. Collectively, these results highlight the importance of PARP family members and ADPRylation in gene regulation, mRNA processing, and protein abundance. Future studies in these areas will yield new insights into the fundamental mechanisms and a broader utility for PARP-targeted therapeutic agents.


ADP-Ribosylation/physiology , Gene Expression/physiology , Poly(ADP-ribose) Polymerases/metabolism , Protein Biosynthesis/physiology , Proteostasis/physiology , Animals , Humans , Protein Processing, Post-Translational , RNA/metabolism
10.
Transl Oncol ; 13(2): 481-489, 2020 Feb.
Article En | MEDLINE | ID: mdl-32004866

Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease with an overall median 5-year survival rate of 8%. This poor prognosis is because of the development of resistance to chemotherapy and radiation therapy and lack of effective targeted therapies. IκB kinase enhancer (IKBKE) overexpression was previously implicated in chemoresistance. Because IKBKE is frequently elevated in PDAC and IKBKE inhibitors are currently in clinical trials, we evaluated IKBKE as a therapeutic target in this disease. Depletion of IKBKE was found to significantly reduce PDAC cell survival, growth, cancer stem cell renewal, and cell migration and invasion. Notably, IKBKE inhibitor CYT387 and IKBKE knockdown dramatically activated the MAPK pathway. Phospho-RTK array analyses showed that IKBKE inhibition leads to rapid upregulation of ErbB3 and IGF-1R expression, which results in MAPK-ERK pathway activation-thereby limiting the efficacy of IKBKE inhibitors. Furthermore, IKBKE inhibition leads to stabilization of FOXO3a, which is required for RTK upregulation on IKBKE inhibition. Finally, we demonstrated that the IKBKE inhibitors synergize with the MEK inhibitor trametinib to significantly induce cell death and inhibit tumor growth and liver metastasis in an orthotopic PDAC mouse model.

11.
Mol Cell ; 75(6): 1270-1285.e14, 2019 09 19.
Article En | MEDLINE | ID: mdl-31351877

PARP inhibitors (PARPi) prevent cancer cell growth by inducing synthetic lethality with DNA repair defects (e.g., in BRCA1/2 mutant cells). We have identified an alternative pathway for PARPi-mediated growth control in BRCA1/2-intact breast cancer cells involving rDNA transcription and ribosome biogenesis. PARP-1 binds to snoRNAs, which stimulate PARP-1 catalytic activity in the nucleolus independent of DNA damage. Activated PARP-1 ADP-ribosylates DDX21, an RNA helicase that localizes to nucleoli and promotes rDNA transcription when ADP-ribosylated. Treatment with PARPi or mutation of the ADP-ribosylation sites reduces DDX21 nucleolar localization, rDNA transcription, ribosome biogenesis, protein translation, and cell growth. The salient features of this pathway are evident in xenografts in mice and human breast cancer patient samples. Elevated levels of PARP-1 and nucleolar DDX21 are associated with cancer-related outcomes. Our studies provide a mechanistic rationale for efficacy of PARPi in cancer cells lacking defects in DNA repair whose growth is inhibited by PARPi.


Breast Neoplasms/metabolism , DEAD-box RNA Helicases/metabolism , Neoplasm Proteins/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , RNA, Neoplasm/metabolism , RNA, Small Nucleolar/metabolism , Ribosomes/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , DEAD-box RNA Helicases/genetics , DNA Repair , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , RNA, Neoplasm/genetics , RNA, Small Nucleolar/genetics , Ribosomes/genetics
12.
Science ; 360(6389)2018 05 11.
Article En | MEDLINE | ID: mdl-29748257

NAD+ (nicotinamide adenine dinucleotide in its oxidized state) is an essential molecule for a variety of physiological processes. It is synthesized in distinct subcellular compartments by three different synthases (NMNAT-1, -2, and -3). We found that compartmentalized NAD+ synthesis by NMNATs integrates glucose metabolism and adipogenic transcription during adipocyte differentiation. Adipogenic signaling rapidly induces cytoplasmic NMNAT-2, which competes with nuclear NMNAT-1 for the common substrate, nicotinamide mononucleotide, leading to a precipitous reduction in nuclear NAD+ levels. This inhibits the catalytic activity of poly[adenosine diphosphate (ADP)-ribose] polymerase-1 (PARP-1), a NAD+-dependent enzyme that represses adipogenic transcription by ADP-ribosylating the adipogenic transcription factor C/EBPß. Reversal of PARP-1-mediated repression by NMNAT-2-mediated nuclear NAD+ depletion in response to adipogenic signals drives adipogenesis. Thus, compartmentalized NAD+ synthesis functions as an integrator of cellular metabolism and signal-dependent transcriptional programs.


Adipocytes/cytology , Adipogenesis/genetics , NAD/biosynthesis , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Transcription, Genetic , Adipocytes/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Glucose/metabolism , Humans , Mice , NAD/genetics , NIH 3T3 Cells , Poly (ADP-Ribose) Polymerase-1/metabolism , Substrate Specificity
13.
J Med Chem ; 60(22): 9290-9298, 2017 11 22.
Article En | MEDLINE | ID: mdl-29111705

Identification of molecular ligands that recognize peptides or proteins is significant but poses a fundamental challenge in chemical biology and biomedical sciences. Development of cyclic peptidomimetic library is scarce, and thus discovery of cyclic peptidomimetic ligands for protein targets is rare. Herein we report the unprecedented one-bead-two-compound (OBTC) combinatorial library based on a novel class of the macrocyclic peptidomimetics γ-AApeptides. In the library, we utilized the coding peptide tags synthesized with Dde-protected α-amino acids, which were orthogonal to solid phase synthesis of γ-AApeptides. Employing the thioether linkage, the desired macrocyclic γ-AApeptides were found to be effective for ligand identification. Screening the library against the receptor tyrosine kinase EphA2 led to the discovery of one lead compound that tightly bound to EphA2 (Kd = 81 nM) and potently antagonized EphA2-mediated signaling. This new approach of macrocyclic peptidomimetic library may lead to a novel platform for biomacromolecular surface recognition and function modulation.


Peptide Library , Peptides, Cyclic/pharmacology , Peptidomimetics/pharmacology , Receptor, EphA2/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Enzyme Assays , Humans , Molecular Dynamics Simulation , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/metabolism , Peptidomimetics/chemical synthesis , Peptidomimetics/metabolism , Protein Binding , Receptor, EphA2/metabolism , Sulfides/chemical synthesis , Sulfides/metabolism , Sulfides/pharmacology
15.
Cancer Res ; 76(15): 4418-29, 2016 08 01.
Article En | MEDLINE | ID: mdl-27287717

Non-small cell lung cancers (NSCLC) marked by EGFR mutations tend to develop resistance to therapeutic EGFR inhibitors, often due to secondary mutation EGFR(T790M) but also other mechanisms. Here we report support for a rationale to target IKBKE, an IκB kinase family member that activates the AKT and NF-κB pathways, as one strategy to address NSCLC resistant to EGFR inhibitors. While wild-type and mutant EGFR directly interacted with IKBKE, only mutant EGFR phosphorylated IKBKE on residues Y153 and Y179. The unphosphorylatable mutant IKBKE-Y153F/Y179-F that lost kinase activity failed to activate AKT and inhibited EGFR signaling. In clinical specimens of NSCLC with activating mutations of EGFR, we observed elevated levels of phospho-Y153 IKBKE. IKBKE ablation with shRNA or small-molecule inhibitor amlexanox selectively inhibited the viability of NSCLC cells with EGFR mutations in vitro In parallel, we found that these treatments activated the MAPK pathway due to attenuation of an IKBKE feedback mechanism. In vivo studies revealed that combining amlexanox with MEK inhibitor AZD6244 significantly inhibited the xenograft tumor growth of NSCLC cells harboring activating EGFR mutations, including EGFR(T790M) Overall, our findings define IKBKE as a direct effector target of EGFR and provide a therapeutic rationale to target IKBKE as a strategy to eradicate EGFR-TKI-resistant NSCLC cells. Cancer Res; 76(15); 4418-29. ©2016 AACR.


Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/metabolism , I-kappa B Kinase/metabolism , Lung Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Lung Neoplasms/pathology , Mutation , Transfection
16.
Ann Allergy Asthma Immunol ; 117(1): 67-71, 2016 07.
Article En | MEDLINE | ID: mdl-27211057

BACKGROUND: A history of a penicillin allergy generally leads to the use of broad-spectrum antibiotics that may increase complications and cost. OBJECTIVE: To determine the cost-effectiveness of performing penicillin skin testing (PST). METHODS: A retrospective analysis was conducted on adult inpatients with a ß-lactam allergy who underwent PST and oral challenge performed by an allergist. The primary outcome was overall antibiotic cost savings for patients switched to a ß-lactam antibiotic (BLA). Secondary outcomes included subsequent admissions that required antibiotics and total number of days a BLA was prescribed. RESULTS: Fifty patients had PST performed (mean age, 62 years). The most common ß-lactam allergy reported was penicillin (92%). Cutaneous reactions were reported in 54% of patients, and 56% had a reaction more than 20 years ago. Fifty percent of patients had aztreonam prescribed before PST. The results of PST were negative in all patients, and 1 patient had anaphylactic symptoms during the oral amoxicillin challenge (98% skin test or oral challenge negative). Thirty-seven patients (75.5%) were changed to a BLA. Overall cost savings were $11,005 ($297 per patient switched to a BLA). There were 31 subsequent admissions that required antibiotics for patients who tested negative on skin test and oral challenge. A BLA was prescribed in 22 of 31 readmissions, totaling 147 days of BLA therapy. CONCLUSION: After the implementation of a PST protocol, we observed a decrease in non-BLA use in patients with previously documented ß-lactam allergy. PST is a safe and cost-effective procedure to serve as a negative predictor test for penicillin hypersensitivity mediated by IgE.


Anti-Bacterial Agents/adverse effects , Drug Hypersensitivity/diagnosis , Hospitalization , Inpatients , Penicillins/adverse effects , Skin Tests , beta-Lactams/adverse effects , Adult , Aged , Anti-Bacterial Agents/economics , Cost-Benefit Analysis , Drug Costs , Drug Hypersensitivity/immunology , Female , Humans , Male , Middle Aged , Phenotype , Retrospective Studies , Skin Tests/methods , Workflow
17.
J Biol Chem ; 290(11): 6857-67, 2015 Mar 13.
Article En | MEDLINE | ID: mdl-25605728

Long noncoding RNAs (lncRNAs) are emerging as key regulators in various biological processes. Epithelial-to-mesenchymal transition (EMT) is a developmental process hijacked by tumor cells to depart from the primary tumor site, invade surrounding tissue, and establish distant metastases. Transforming growth factor ß (TGFß) signaling has been shown to be a major inducer of EMT and to facilitate breast cancer metastasis. However, the role of lncRNAs in this process remains largely unknown. Here we report a genome-wide lncRNA profile in mouse mammary epithelial NMuMG cells upon TGFß induction of EMT. Among 10,802 lncRNAs profiled, over 600 were up-regulated and down-regulated during the EMT, respectively. Furthermore, we identify that lncRNA-HIT (HOXA transcript induced by TGFß) mediates TGFß function, i.e. depletion of lncRNA-HIT inhibits TGFß-induced migration, invasion, and EMT in NMuMG. LncRNA-HIT is also significantly elevated in the highly metastatic 4T1 cells. Knockdown of lncRNA-HIT in 4T1 results in decrease of cell migration, invasion, tumor growth, and metastasis. E-cadherin was identified as a major target of lncRNA-HIT. Moreover, lncRNA-HIT is conserved in humans and elevated expression associates with more invasive human primary breast carcinoma. Collectively, these data suggest that a subset of lncRNAs such as lncRNA-HIT play a significant role in regulation of EMT and breast cancer invasion and metastasis, and could be potential therapeutic targets in breast cancers.


Breast Neoplasms/pathology , Breast/pathology , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Neoplasm Invasiveness/genetics , RNA, Long Noncoding/genetics , Transforming Growth Factor beta/metabolism , Animals , Breast/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Line , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lung/metabolism , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , Neoplasm Invasiveness/pathology , RNA, Long Noncoding/metabolism , Transcriptome
18.
J Biol Chem ; 288(52): 36971-82, 2013 Dec 27.
Article En | MEDLINE | ID: mdl-24235142

Transforming growth factor ß (TGFß) is a potent and context-dependent regulator of tumor progression. TGFß promotes the lung metastasis of basal-like (but not the luminal-like) breast cancer. Here, we demonstrated that fascin, a pro-metastasis actin bundling protein, was a direct target of the canonical TGFß-Smad4 signaling pathway in basal-like breast cancer cells. TGFß and Smad4 induced fascin overexpression by directly binding to a Smad binding element on the fascin promoter. We identified GATA3, a transcription factor crucial for mammary gland morphogenesis and luminal differentiation, as a negative regulator of TGFß- and Smad4-induced fascin overexpression. When ectopically expressed in basal-like breast cancer cells, GATA-3 abrogated TGFß- and Smad4-mediated overexpression of fascin and other TGFß response genes, invadopodium formation, cell migration, and invasion, suggesting suppression of the canonical TGFß-Smad signaling axis. Mechanistically, GATA3 abrogated the canonical TGFß-Smad signaling by abolishing interactions between Smad4 and its DNA binding elements, potentially through physical interactions between the N-terminal of GATA3 and Smad3/4 proteins. Our findings provide mechanistic insight into how TGFß-mediated cell motility and invasiveness are differentially regulated in breast cancer.


Breast Neoplasms/metabolism , Carrier Proteins/biosynthesis , Cell Movement , GATA3 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Microfilament Proteins/biosynthesis , Neoplasm Proteins/metabolism , Smad4 Protein/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carrier Proteins/genetics , Cell Line, Tumor , Female , GATA3 Transcription Factor/genetics , HEK293 Cells , Humans , Microfilament Proteins/genetics , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Response Elements , Signal Transduction/genetics , Smad3 Protein/genetics , Smad3 Protein/metabolism , Smad4 Protein/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
19.
J Card Fail ; 19(2): 101-7, 2013 Feb.
Article En | MEDLINE | ID: mdl-23384635

BACKGROUND: An impaired cardiac output response to exercise is a hallmark of chronic heart failure (HF). We determined the extent to which noninvasive estimates of cardiac hemodynamics during exercise in combination with cardiopulmonary exercise test (CPX) responses improved the estimation of risk for adverse events in patients with HF. METHODS AND RESULTS: CPX and impedance cardiography were performed in 639 consecutive patients (mean age 48 ± 14 years), evaluated for HF. Clinical, hemodynamic, and CPX variables were acquired at baseline and subjects were followed for a mean of 460 ± 332 days. Patients were followed for the composite outcome of cardiac-related death, hospitalization for worsening HF, cardiac transplantation, and left ventricular assist device implantation. Cox proportional hazards analyses including clinical, noninvasive hemodynamic, and CPX variables were performed to determine their association with the composite endpoint. There were 113 events. Among CPX variables, peak oxygen uptake (VO(2)) and the minute ventilation (VE)/carbon dioxide production (VCO(2)) slope were significant predictors of risk for adverse events (age-adjusted hazard ratio [HR] 1.08, 95% confidence interval [CI] 1.05-1.11 for both; P < .001). Among hemodynamic variables, peak cardiac index was the strongest predictor of risk (HR 1.08, 95% CI 1.0-1.16; P = .01). In a multivariate analysis including CPX and noninvasively determined hemodynamic variables, the most powerful predictive model included the combination of peak VO(2), peak cardiac index, and the VE/VCO(2) slope, with each contributing significantly and independently to predicting risk; an abnormal response for all 3 yielded an HR of 5.1 (P < .001). CONCLUSIONS: These findings suggest that noninvasive indices of cardiac hemodynamics complement established CPX measures in quantifying risk in patients with HF.


Exercise Test/methods , Exercise/physiology , Heart Failure/diagnosis , Heart Failure/physiopathology , Hemodynamics/physiology , Adult , Aged , Female , Heart Failure/therapy , Humans , Male , Middle Aged , Predictive Value of Tests , Treatment Outcome
20.
Prostate ; 70(12): 1274-85, 2010 Sep 01.
Article En | MEDLINE | ID: mdl-20623637

BACKGROUND: Androgen receptor (AR) plays a critical role in the progression of both androgen-dependent and androgen-independent prostate cancer (AIPC). Ligand-independent activation of AR in AIPC or castration resistant prostate cancer (CRPC) is often associated with poor prognosis. Recently, tyrosine kinase Ack1 has been shown to regulate AR activity by phosphorylating it at tyrosine 267 and this event was shown to be critical for AIPC growth. However, whether a small molecule inhibitor that can mitigate Ack1 activation is sufficient to abrogate AR activity on AR regulated promoters in androgen-depleted environment is not known. METHODS: We have generated two key resources, antibodies that specifically recognize pTyr267-AR and synthesized a small molecule inhibitor of Ack1, 4-amino-5,6-biaryl-furo[2,3-d]pyrimidine (named here as AIM-100) to test whether AIM-100 modulates ligand-independent AR activity and inhibits prostate cell growth. RESULTS: Prostate tissue microarray analysis indicates that Ack1 Tyr284 phosphorylation correlates positively with disease progression and negatively with the survival of prostate cancer patients. Interestingly, neither pTyr267-AR expression nor its transcriptional activation was affected by anti-androgens in activated Ack1 expressing or EGF stimulated prostate cells. However, the Ack1 inhibitor, AIM-100, not only inhibited Ack1 activation but also able to suppress pTyr267-AR phosphorylation, binding of AR to PSA, NKX3.1, and TMPRSS2 promoters, and inhibit AR transcription activity. CONCLUSION: Ack1 Tyr284 phosphorylation is prognostic of progression of prostate cancer and inhibitors of Ack1 activity could be novel therapeutic agents to treat AIPC.


Androgens/physiology , Prostatic Neoplasms/physiopathology , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Androgen/physiology , Amino Acid Sequence , Androgens/genetics , Animals , Antibody Formation , Cell Line, Tumor , Disease Progression , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Male , Oligonucleotide Array Sequence Analysis , Phosphopeptides/chemistry , Prostatic Hyperplasia/complications , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Rabbits , Receptors, Androgen/genetics , Receptors, Androgen/immunology , Survival Rate
...