Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
BMC Cancer ; 24(1): 527, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664630

ABSTRACT

BACKGROUND: Tumor Treating Fields (TTFields) Therapy is an FDA-approved therapy in the first line and recurrent setting for glioblastoma. Despite Phase 3 evidence showing improved survival with TTFields, it is not uniformly utilized. We aimed to examine patient and clinician views of TTFields and factors shaping utilization of TTFields through a unique research partnership with medical neuro oncology and medical social sciences. METHODS: Adult glioblastoma patients who were offered TTFields at a tertiary care academic hospital were invited to participate in a semi-structured interview about their decision to use or not use TTFields. Clinicians who prescribe TTFields were invited to participate in a semi-structured interview about TTFields. RESULTS: Interviews were completed with 40 patients with a mean age of 53 years; 92.5% were white and 60% were male. Participants who decided against TTFields stated that head shaving, appearing sick, and inconvenience of wearing/carrying the device most influenced their decision. The most influential factors for use of TTFields were the efficacy of the device and their clinician's opinion. Clinicians (N = 9) stated that TTFields was a good option for glioblastoma patients, but some noted that their patients should consider the burdens and benefits of TTFields as it may not be the desired choice for all patients. CONCLUSIONS: This is the first study to examine patient decision making for TTFields. Findings suggest that clinician support and efficacy data are among the key decision-making factors. Properly understanding the path to patients' decision making is crucial in optimizing the use of TTFields and other therapeutic decisions for glioblastoma patients.


Subject(s)
Brain Neoplasms , Decision Making , Glioblastoma , Humans , Male , Middle Aged , Brain Neoplasms/therapy , Female , Glioblastoma/therapy , Adult , Aged , Electric Stimulation Therapy/methods , Qualitative Research , Physicians/psychology , Clinical Decision-Making
2.
Lancet Oncol ; 24(5): 509-522, 2023 05.
Article in English | MEDLINE | ID: mdl-37142373

ABSTRACT

BACKGROUND: Low-intensity pulsed ultrasound with concomitant administration of intravenous microbubbles (LIPU-MB) can be used to open the blood-brain barrier. We aimed to assess the safety and pharmacokinetics of LIPU-MB to enhance the delivery of albumin-bound paclitaxel to the peritumoural brain of patients with recurrent glioblastoma. METHODS: We conducted a dose-escalation phase 1 clinical trial in adults (aged ≥18 years) with recurrent glioblastoma, a tumour diameter of 70 mm or smaller, and a Karnofsky performance status of at least 70. A nine-emitter ultrasound device was implanted into a skull window after tumour resection. LIPU-MB with intravenous albumin-bound paclitaxel infusion was done every 3 weeks for up to six cycles. Six dose levels of albumin-bound paclitaxel (40 mg/m2, 80 mg/m2, 135 mg/m2, 175 mg/m2, 215 mg/m2, and 260 mg/m2) were evaluated. The primary endpoint was dose-limiting toxicity occurring during the first cycle of sonication and albumin-bound paclitaxel chemotherapy. Safety was assessed in all treated patients. Analyses were done in the per-protocol population. Blood-brain barrier opening was investigated by MRI before and after sonication. We also did pharmacokinetic analyses of LIPU-MB in a subgroup of patients from the current study and a subgroup of patients who received carboplatin as part of a similar trial (NCT03744026). This study is registered with ClinicalTrials.gov, NCT04528680, and a phase 2 trial is currently open for accrual. FINDINGS: 17 patients (nine men and eight women) were enrolled between Oct 29, 2020, and Feb 21, 2022. As of data cutoff on Sept 6, 2022, median follow-up was 11·89 months (IQR 11·12-12·78). One patient was treated per dose level of albumin-bound paclitaxel for levels 1 to 5 (40-215 mg/m2), and 12 patients were treated at dose level 6 (260 mg/m2). A total of 68 cycles of LIPU-MB-based blood-brain barrier opening were done (median 3 cycles per patient [range 2-6]). At a dose of 260 mg/m2, encephalopathy (grade 3) occurred in one (8%) of 12 patients during the first cycle (considered a dose-limiting toxicity), and in one other patient during the second cycle (grade 2). In both cases, the toxicity resolved and treatment continued at a lower dose of albumin-bound paclitaxel, with a dose of 175 mg/m2 in the case of the grade 3 encephalopathy, and to 215 mg/m2 in the case of the grade 2 encephalopathy. Grade 2 peripheral neuropathy was observed in one patient during the third cycle of 260 mg/m2 albumin-bound paclitaxel. No progressive neurological deficits attributed to LIPU-MB were observed. LIPU-MB-based blood-brain barrier opening was most commonly associated with immediate yet transient grade 1-2 headache (12 [71%] of 17 patients). The most common grade 3-4 treatment-emergent adverse events were neutropenia (eight [47%]), leukopenia (five [29%]), and hypertension (five [29%]). No treatment-related deaths occurred during the study. Imaging analysis showed blood-brain barrier opening in the brain regions targeted by LIPU-MB, which diminished over the first 1 h after sonication. Pharmacokinetic analyses showed that LIPU-MB led to increases in the mean brain parenchymal concentrations of albumin-bound paclitaxel (from 0·037 µM [95% CI 0·022-0·063] in non-sonicated brain to 0·139 µM [0·083-0·232] in sonicated brain [3·7-times increase], p<0·0001) and carboplatin (from 0·991 µM [0·562-1·747] in non-sonicated brain to 5·878 µM [3·462-9·980] µM in sonicated brain [5·9-times increase], p=0·0001). INTERPRETATION: LIPU-MB using a skull-implantable ultrasound device transiently opens the blood-brain barrier allowing for safe, repeated penetration of cytotoxic drugs into the brain. This study has prompted a subsequent phase 2 study combining LIPU-MB with albumin-bound paclitaxel plus carboplatin (NCT04528680), which is ongoing. FUNDING: National Institutes of Health and National Cancer Institute, Moceri Family Foundation, and the Panattoni family.


Subject(s)
Brain Diseases , Glioblastoma , Adult , Male , Humans , Female , Adolescent , Albumin-Bound Paclitaxel/adverse effects , Carboplatin , Glioblastoma/diagnostic imaging , Glioblastoma/drug therapy , Blood-Brain Barrier , Paclitaxel , Brain Diseases/chemically induced , Brain Diseases/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
3.
Chem Commun (Camb) ; 59(42): 6347-6350, 2023 May 23.
Article in English | MEDLINE | ID: mdl-37144553

ABSTRACT

To ensure specificity of small interfering RNAs (siRNAs), the antisense strand must be selected by the RNA-induced silencing complex (RISC). We have previously demonstrated that a 5'-morpholino-modified nucleotide at the 5'-end of the sense strand inhibits its interaction with RISC ensuring selection of the desired antisense strand. To improve this antagonizing binding property even further, a new set of morpholino-based analogues, Mo2 and Mo3, and a piperidine analogue, Pip, were designed based on the known structure of Argonaute2, the slicer enzyme component of RISC. Sense strands of siRNAs were modified with these new analogues, and the siRNAs were evaluated in vitro and in mice for RNAi activity. Our data demonstrated that Mo2 is the best RISC inhibitor among the modifications tested and that it effectively mitigates sense strand-based off-target activity of siRNA.


Subject(s)
RNA, Small Interfering , RNA-Induced Silencing Complex , Animals , Mice , RNA, Small Interfering/chemistry , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism , Morpholinos/chemistry
4.
Nat Commun ; 14(1): 1970, 2023 04 08.
Article in English | MEDLINE | ID: mdl-37031257

ABSTRACT

Adeno-associated virus (AAV)-based gene therapy could be facilitated by the development of molecular switches to control the magnitude and timing of expression of therapeutic transgenes. RNA interference (RNAi)-based approaches hold unique potential as a clinically proven modality to pharmacologically regulate AAV gene dosage in a sequence-specific manner. We present a generalizable RNAi-based rheostat wherein hepatocyte-directed AAV transgene expression is silenced using the clinically validated modality of chemically modified small interfering RNA (siRNA) conjugates or vectorized co-expression of short hairpin RNA (shRNA). For transgene induction, we employ REVERSIR technology, a synthetic high-affinity oligonucleotide complementary to the siRNA or shRNA guide strand to reverse RNAi activity and rapidly recover transgene expression. For potential clinical development, we report potent and specific siRNA sequences that may allow selective regulation of transgenes while minimizing unintended off-target effects. Our results establish a conceptual framework for RNAi-based regulatory switches with potential for infrequent dosing in clinical settings to dynamically modulate expression of virally-delivered gene therapies.


Subject(s)
Dependovirus , Genetic Therapy , RNA Interference , Dependovirus/genetics , Dependovirus/metabolism , RNA, Small Interfering/metabolism , Transgenes , RNA, Double-Stranded , Genetic Vectors/genetics
5.
Drug Metab Dispos ; 50(6): 781-797, 2022 06.
Article in English | MEDLINE | ID: mdl-34154993

ABSTRACT

Conjugation of oligonucleotide therapeutics, including small interfering RNAs (siRNAs) or antisense oligonucleotides, to N-acetylgalactosamine (GalNAc) ligands has become the primary strategy for hepatocyte-targeted delivery, and with the recent approvals of GIVLAARI (givosiran) for the treatment of acute hepatic porphyria, OXLUMO (lumasiran) for the treatment of primary hyperoxaluria, and Leqvio (inclisiran) for the treatment of hypercholesterolemia, the technology has been well validated clinically. Although much knowledge has been gained over decades of development, there is a paucity of published literature on the drug metabolism and pharmacokinetic properties of GalNAc-siRNA. With this in mind, the goals of this minireview are to provide an aggregate analysis of these nonclinical absorption, distribution, metabolism, and excretion (ADME) data to build confidence on the translation of these properties to human. Upon subcutaneous administration, GalNAc-conjugated siRNAs are quickly distributed to the liver, resulting in plasma pharmacokinetic (PK) properties that reflect rapid elimination through asialoglycoprotein receptor-mediated uptake from circulation into hepatocytes. These studies confirm that liver PK, including half-life and, most importantly, siRNA levels in RNA-induced silencing complex in hepatocytes, are better predictors of pharmacodynamics (PD) than plasma PK. Several in vitro and in vivo nonclinical studies were conducted to characterize the ADME properties of GalNAc-conjugated siRNAs. These studies demonstrate that the PK/PD and ADME properties of GalNAc-conjugated siRNAs are highly conserved across species, are largely predictable, and can be accurately scaled to human, allowing us to identify efficacious and safe clinical dosing regimens in the absence of human liver PK profiles. SIGNIFICANCE STATEMENT: Several nonclinical ADME studies have been conducted in order to provide a comprehensive overview of the disposition and elimination of GalNAc-conjugated siRNAs and the pharmacokinetic/pharmacodynamic translation between species. These studies demonstrate that the ADME properties of GalNAc-conjugated siRNAs are well correlated and predictable across species, building confidence in the ability to extrapolate to human.


Subject(s)
Acetylgalactosamine , Porphyrias, Hepatic , Acetylgalactosamine/pharmacokinetics , Asialoglycoprotein Receptor/metabolism , Hepatocytes/metabolism , Humans , Porphyrias, Hepatic/metabolism , RNA, Small Interfering/genetics
6.
Nucleic Acids Res ; 48(21): 11827-11844, 2020 12 02.
Article in English | MEDLINE | ID: mdl-32808038

ABSTRACT

One hallmark of trivalent N-acetylgalactosamine (GalNAc)-conjugated siRNAs is the remarkable durability of silencing that can persist for months in preclinical species and humans. Here, we investigated the underlying biology supporting this extended duration of pharmacological activity. We found that siRNA accumulation and stability in acidic intracellular compartments is critical for long-term activity. We show that functional siRNA can be liberated from these compartments and loaded into newly generated Argonaute 2 protein complexes weeks after dosing, enabling continuous RNAi activity over time. Identical siRNAs delivered in lipid nanoparticles or as GalNAc conjugates were dose-adjusted to achieve similar knockdown, but only GalNAc-siRNAs supported an extended duration of activity, illustrating the importance of receptor-mediated siRNA trafficking in the process. Taken together, we provide several lines of evidence that acidic intracellular compartments serve as a long-term depot for GalNAc-siRNA conjugates and are the major contributor to the extended duration of activity observed in vivo.


Subject(s)
Acetylgalactosamine/metabolism , Asialoglycoprotein Receptor/metabolism , Drug Carriers , Gene Silencing , Prealbumin/genetics , RNA, Small Interfering/metabolism , Acetylgalactosamine/chemistry , Animals , Argonaute Proteins/genetics , Asialoglycoprotein Receptor/genetics , Biological Transport , Drug Stability , Female , Glycoconjugates/chemistry , Glycoconjugates/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Hydrogen-Ion Concentration , Liver/cytology , Liver/metabolism , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Nanoparticles/metabolism , Prealbumin/antagonists & inhibitors , Prealbumin/metabolism , RNA, Small Interfering/genetics , Time Factors
7.
Cell Rep ; 30(2): 541-554.e5, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31940495

ABSTRACT

Long non-coding RNAs (lncRNAs) are critical regulators of numerous physiological processes and diseases, especially cancers. However, development of lncRNA-based therapies is limited because the mechanisms of many lncRNAs are obscure, and interactions with functional partners, including proteins, remain uncharacterized. The lncRNA SLNCR1 binds to and regulates the androgen receptor (AR) to mediate melanoma invasion and proliferation in an androgen-independent manner. Here, we use biochemical analyses coupled with selective 2'-hydroxyl acylation analyzed by primer extension (SHAPE) RNA structure probing to show that the N-terminal domain of AR binds a pyrimidine-rich motif in an unstructured region of SLNCR1. This motif is predictive of AR binding, as we identify an AR-binding motif in lncRNA HOXA11-AS-203. Oligonucleotides that bind either the AR N-terminal domain or the AR RNA motif block the SLNCR1-AR interaction and reduce SLNCR1-mediated melanoma invasion. Delivery of oligos that block SLNCR1-AR interaction thus represent a plausible therapeutic strategy.


Subject(s)
Melanoma/metabolism , RNA, Long Noncoding/metabolism , Receptors, Androgen/metabolism , Base Sequence , Cell Line, Tumor , Cell Proliferation/physiology , Female , HEK293 Cells , Humans , Male , Melanoma/genetics , Melanoma/pathology , Neoplasm Invasiveness , Protein Domains , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptors, Androgen/genetics
8.
Cell Rep ; 27(8): 2493-2507.e4, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31116991

ABSTRACT

Melanoma is the deadliest form of skin cancer, affecting men more frequently and severely than women. Although recent studies suggest that differences in activity of the androgen receptor (AR) underlie the observed sex bias, little is known about AR activity in melanoma. Here we show that AR and EGR1 bind to the long non-coding RNA SLNCR and increase melanoma proliferation through coordinated transcriptional regulation of several growth-regulatory genes. ChIP-seq reveals that ligand-free AR is enriched on SLNCR-regulated melanoma genes and that AR genomic occupancy significantly overlaps with EGR1 at consensus EGR1 binding sites. We present a model in which SLNCR recruits AR to EGR1-bound genomic loci and switches EGR1-mediated transcriptional activation to repression of the tumor suppressor p21Waf1/Cip1. Our data implicate the regulatory triad of SLNCR, AR, and EGR1 in promoting oncogenesis and may help explain why men have a higher incidence of and more rapidly progressive melanomas compared with women.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Early Growth Response Protein 1/metabolism , RNA, Long Noncoding/metabolism , Receptors, Androgen/metabolism , Binding Sites , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Early Growth Response Protein 1/chemistry , Female , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Humans , Ligands , Male , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Protein Binding , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism
9.
J Biol Methods ; 4(1)2017.
Article in English | MEDLINE | ID: mdl-29201933

ABSTRACT

Long non-coding RNAs (lncRNAs) regulate critical cellular processes and their dysregulation contributes to multiple diseases. Although only a few lncRNAs have defined mechanisms, many of these characterized lncRNAs interact with transcription factors to regulate gene expression, suggesting a common mechanism of action. Identifying RNA-bound transcription factors is especially challenging due to inefficient RNA immunoprecipitation and low abundance of many transcription factors. Here we describe a highly sensitive, user-friendly, and inexpensive technique called RATA (RNA-associated transcription factor array), which utilizes a MS2-aptamer pulldown strategy coupled with transcription factor activation arrays for identification of transcription factors associated with a nuclear RNA of interest. RATA requires only ~5 million cells and standard molecular biology reagents for multiplexed identification of up to 96 transcription factors in 2-3 d. Thus, RATA offers significant advantages over other technologies for analysis of RNA-transcription factor interactions.

10.
Cell Rep ; 15(9): 2025-37, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27210747

ABSTRACT

Long non-coding RNAs (lncRNAs) have been implicated in numerous physiological processes and diseases, most notably cancers. However, little is known about the mechanism of many functional lncRNAs. We identified an abundantly expressed lncRNA associated with decreased melanoma patient survival. Increased expression of this lncRNA, SLNCR1, mediates melanoma invasion through a highly conserved sequence similar to that of the lncRNA SRA1. Using a sensitive technique we term RATA (RNA-associated transcription factor array), we show that the brain-specific homeobox protein 3a (Brn3a) and the androgen receptor (AR) bind within and adjacent to SLNCR1's conserved region, respectively. SLNCR1, AR, and Brn3a are specifically required for transcriptional activation of matrix metalloproteinase 9 (MMP9) and increased melanoma invasion. Our observations directly link AR to melanoma invasion, possibly explaining why males experience more melanoma metastases and have an overall lower survival in comparison to females.


Subject(s)
Conserved Sequence/genetics , Melanoma/genetics , Melanoma/pathology , RNA, Long Noncoding/genetics , Base Sequence , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Models, Biological , Neoplasm Invasiveness , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Receptors, Androgen , Survival Analysis , Transcription Factor Brn-3A/metabolism , Transcription, Genetic , Up-Regulation/genetics
11.
PLoS One ; 9(11): e112921, 2014.
Article in English | MEDLINE | ID: mdl-25391136

ABSTRACT

Bacteria have evolved mechanisms that allow them to survive in the face of a variety of stresses including nutrient deprivation, antibiotic challenge and engulfment by predator cells. A switch to dormancy represents one strategy that reduces energy utilization and can render cells resistant to compounds that kill growing bacteria. These persister cells pose a problem during treatment of infections with antibiotics, and dormancy mechanisms may contribute to latent infections. Many bacteria encode toxin-antitoxin (TA) gene pairs that play an important role in dormancy and the formation of persisters. VapBC gene pairs comprise the largest of the Type II TA systems in bacteria and they produce a VapC ribonuclease toxin whose activity is inhibited by the VapB antitoxin. Despite the importance of VapBC TA pairs in dormancy and persister formation, little information exists on the structural features of VapC proteins required for their toxic function in vivo. Studies reported here identified 17 single mutations that disrupt the function of VapC1 from non-typeable H. influenzae in vivo. 3-D modeling suggests that side chains affected by many of these mutations sit near the active site of the toxin protein. Phylogenetic comparisons and secondary mutagenesis indicate that VapC1 toxicity requires an alternative active site motif found in many proteobacteria. Expression of the antitoxin VapB1 counteracts the activity of VapC1 mutants partially defective for toxicity, indicating that the antitoxin binds these mutant proteins in vivo. These findings identify critical chemical features required for the biological function of VapC toxins and PIN-domain proteins.


Subject(s)
Antitoxins/genetics , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Catalytic Domain/genetics , Haemophilus influenzae/genetics , Mutation/genetics , Amino Acid Sequence , Gene Expression/genetics , Molecular Sequence Data , Protein Structure, Tertiary/genetics , Ribonucleases/genetics , Sequence Alignment
12.
Wiley Interdiscip Rev RNA ; 4(2): 217-31, 2013.
Article in English | MEDLINE | ID: mdl-23417976

ABSTRACT

The advent of high-throughput sequencing technologies has revealed that pervasive transcription generates RNAs from nearly all regions of eukaryotic genomes. Normally, these transcripts undergo rapid degradation by a nuclear RNA surveillance system primarily featuring the RNA exosome. This multimeric protein complex plays a critical role in the efficient turnover and processing of a vast array of RNAs in the nucleus. Despite its initial discovery over a decade ago, important questions remain concerning the mechanisms that recruit and activate the nuclear exosome. Specificity and modulation of exosome activity requires additional protein cofactors, including the conserved TRAMP polyadenylation complex. Recent studies suggest that helicase and RNA-binding subunits of TRAMP direct RNA substrates for polyadenylation, which enhances their degradation by Dis3/Rrp44 and Rrp6, the two exosome-associated ribonucleases. These findings indicate that the exosome and TRAMP have evolved highly flexible functions that allow recognition of a wide range of RNA substrates. This flexibility provides the nuclear RNA surveillance system with the ability to regulate the levels of a broad range of coding and noncoding RNAs, which results in profound effects on gene expression, cellular development, gene silencing, and heterochromatin formation. This review summarizes recent findings on the nuclear RNA surveillance complexes, and speculates upon possible mechanisms for TRAMP-mediated substrate recognition and exosome activation.


Subject(s)
Exosomes/metabolism , Multiprotein Complexes/metabolism , RNA, Nuclear/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Exosomes/genetics , Humans , Multiprotein Complexes/genetics , RNA, Nuclear/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Species Specificity
13.
RNA ; 18(10): 1934-45, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22923767

ABSTRACT

RNA surveillance systems function at critical steps during the formation and function of RNA molecules in all organisms. The RNA exosome plays a central role in RNA surveillance by processing and degrading RNA molecules in the nucleus and cytoplasm of eukaryotic cells. The exosome functions as a complex of proteins composed of a nine-member core and two ribonucleases. The identity of the molecular determinants of exosome RNA substrate specificity remains an important unsolved aspect of RNA surveillance. In the nucleus of Saccharomyces cerevisiae, TRAMP complexes recognize and polyadenylate RNAs, which enhances RNA degradation by the exosome and may contribute to its specificity. TRAMPs contain either of two putative RNA-binding factors called Air proteins. Previous studies suggested that these proteins function interchangeably in targeting the poly(A)-polymerase activity of TRAMPs to RNAs. Experiments reported here show that the Air proteins govern separable functions. Phenotypic analysis and RNA deep-sequencing results from air mutants reveal specific requirements for each Air protein in the regulation of the levels of noncoding and coding RNAs. Loss of these regulatory functions results in specific metabolic and plasmid inheritance defects. These findings reveal differential functions for Air proteins in RNA metabolism and indicate that they control the substrate specificity of the RNA exosome.


Subject(s)
RNA Stability , RNA, Nuclear/metabolism , RNA-Binding Proteins/physiology , Saccharomyces cerevisiae Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Cell Nucleus/genetics , Cell Nucleus/metabolism , Exosome Multienzyme Ribonuclease Complex/genetics , Exosome Multienzyme Ribonuclease Complex/metabolism , High-Throughput Nucleotide Sequencing , Organisms, Genetically Modified , Poly A/metabolism , Protein Binding , RNA Stability/genetics , RNA Stability/physiology , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Substrate Specificity
14.
J Crit Care ; 27(3): 321.e5-12, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22425340

ABSTRACT

PURPOSE: Multiple equations exist for predicting resting energy expenditure (REE). The accuracy of these for estimating energy requirements of critically ill patients is not clear, especially for obese patients. We sought to compare REE, calculated with published formulas, with measured REE in a cohort of mechanically ventilated subjects. MATERIALS AND METHODS: We retrospectively identified all mechanically ventilated patients with measured body mass index who underwent indirect calorimetry in the intensive care unit. Limits of agreement and Pitman's test of difference in variance were performed to compare REE by equations with REE measured by indirect calorimetry. RESULTS: A total of 927 patients were identified, including 401 obese patients. There were bias and poor agreement between measured REE and REE predicted by the Harris-Benedict, Owen, American College of Chest Physicians, and Mifflin equations (P > .05). There was poor agreement between measured and predicted REE by the Ireton-Jones equation, stratifying by sex. Ireton-Jones was the only equation that was unbiased for men and those in weight categories 1 and 2. In all cases except Ireton-Jones, predictive equations underestimated measured REE. CONCLUSION: None of these equations accurately estimated measured REE in this group of mechanically ventilated patients, most underestimating energy needs. Development of improved predictive equations for adequate assessment of energy needs is needed.


Subject(s)
Critical Illness , Decision Support Techniques , Energy Metabolism , Nutritional Requirements , Obesity/physiopathology , Respiration, Artificial , Body Mass Index , Calorimetry, Indirect , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Reproducibility of Results , Retrospective Studies , Washington
15.
Biochem Biophys Res Commun ; 383(2): 198-202, 2009 May 29.
Article in English | MEDLINE | ID: mdl-19345671

ABSTRACT

The Saccharomyces cerevisiae gene YPK9 encodes a putative integral membrane protein which is 58% similar and 38% identical in amino acid sequence to the human lysosomal P(5B) ATPase ATP13A2. Mutations in ATP13A2 have been found in patients with Kufor-Rakeb syndrome, a form of juvenile Parkinsonism. We report that Ypk9p localizes to the yeast vacuole and that deletion of YPK9 confers sensitivity for growth for cadmium, manganese, nickel or selenium. These results suggest that Ypk9p may play a role in sequestration of divalent heavy metal ions. Further studies on the function of Ypk9p/ATP13A2 may help to define the molecular basis of Kufor-Rakeb syndrome and provide a potential link to environmental factors such as heavy metals contributing to some forms of Parkinsonism.


Subject(s)
Cadmium/toxicity , Manganese/toxicity , Nickel/toxicity , Proton-Translocating ATPases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/drug effects , Selenium/toxicity , Cadmium/metabolism , Cations, Divalent/metabolism , Cations, Divalent/toxicity , Histidine/metabolism , Humans , Manganese/metabolism , Nickel/metabolism , Proton-Translocating ATPases/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Selenium/metabolism , Vacuoles/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL