Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Article in English | MEDLINE | ID: mdl-38349120

ABSTRACT

It is unclear what effect biological sex has on the outcomes of acute lung injury (ALI). Clinical studies are confounded by their observational design. We addressed this knowledge gap with a preclinical systematic review of ALI animal studies. We searched MEDLINE and Embase for studies of intratracheal/intranasal/aerosolized lipopolysaccharide (LPS) administration, the most common ALI model, and reported sex-stratified data. Screening and data extraction were conducted in duplicate. Our primary outcome was histological tissue injury and secondary outcomes included alveolar-capillary barrier alterations and inflammatory markers. We used a random effects inverse variance meta-analysis, expressing data as standardized mean difference (SMD) with 95% confidence intervals (CI). Risk of bias was assessed using the SYRCLE tool. We identified six studies involving 132 animals across 11 independent experiments. A total of 41 outcomes were extracted, with the direction of effect suggesting greater severity in males than females in 26/41 outcomes (63%). One study reported on lung histology and found that male mice exhibited greater injury than females (SMD 1.61, 95% CI 0.53 to 2.69). Meta-analysis demonstrated significantly elevated albumin levels (SMD 2.17, 95% CI 0.63 to 3.70) and total cell counts (SMD 0.80, 95% CI 0.27 to 1.33) in bronchoalveolar lavage fluid from male mice compared to females. Most studies had an 'unclear risk of bias'. Our findings suggest sex-related differences in ALI severity. However, these conclusions are drawn from a small number of animals and studies. Further research is required to address the fundamental issue of biological sex differences in LPS-induced ALI.

2.
Adv Sci (Weinh) ; 10(26): e2302611, 2023 09.
Article in English | MEDLINE | ID: mdl-37400371

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a rare disease involving cystic lung destruction by invasive LAM cells. These cells harbor loss-of-function mutations in TSC2, conferring hyperactive mTORC1 signaling. Here, tissue engineering tools are employed to model LAM and identify new therapeutic candidates. Biomimetic hydrogel culture of LAM cells is found to recapitulate the molecular and phenotypic characteristics of human disease more faithfully than culture on plastic. A 3D drug screen is conducted, identifying histone deacetylase (HDAC) inhibitors as anti-invasive agents that are also selectively cytotoxic toward TSC2-/- cells. The anti-invasive effects of HDAC inhibitors are independent of genotype, while selective cell death is mTORC1-dependent and mediated by apoptosis. Genotype-selective cytotoxicity is seen exclusively in hydrogel culture due to potentiated differential mTORC1 signaling, a feature that is abrogated in cell culture on plastic. Importantly, HDAC inhibitors block invasion and selectively eradicate LAM cells in vivo in zebrafish xenografts. These findings demonstrate that tissue-engineered disease modeling exposes a physiologically relevant therapeutic vulnerability that would be otherwise missed by conventional culture on plastic. This work substantiates HDAC inhibitors as possible therapeutic candidates for the treatment of patients with LAM and requires further study.


Subject(s)
Lung Neoplasms , Lymphangioleiomyomatosis , Animals , Humans , Lymphangioleiomyomatosis/drug therapy , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/metabolism , Lung Neoplasms/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Tissue Engineering , Zebrafish , Mechanistic Target of Rapamycin Complex 1
3.
J Crit Care ; 75: 154284, 2023 06.
Article in English | MEDLINE | ID: mdl-36870801

ABSTRACT

INTRODUCTION: We aimed to analyze intensive care unit (ICU)-acquired pneumonia according to 7 definitions, estimating associated hospital mortality. METHODS: This cohort study was nested within an international randomized trial, evaluating the effect of probiotics on ICU-acquired pneumonia in 2650 mechanically ventilated adults. Each clinically suspected pneumonia was adjudicated by two physicians blinded to allocation and center. The primary outcome was ventilator-associated pneumonia (VAP) informed by ventilation for ≥2 days, new, progressive or persistent infiltrate plus 2 of: temperature > 38 °C or < 36 °C; leukopenia (<3 × 10(Fernando et al., 20206)/L) or leukocytosis (>10 × 10(Fernando et al., 20206)/L); and purulent sputum. We also used 6 other definitions estimating the risk of hospital mortality. RESULTS: The frequency of ICU-acquired pneumonia varied by definition: the trial primary outcome VAP (21.6%), Clinical Pulmonary Infection Score (CPIS) (24.9%), American College Chest Physicians (ACCP) (25.0%), International Sepsis Forum (ISF) (24.4%), Reducing Oxidative Stress Study (REDOXS) (17.6%), Centers for Disease Control (CDC) (7.8%), and invasively microbiologically confirmed (1.9%). The trial primary outcome VAP (HR 1.31 [1.08, 1.60]), ISF (HR 1.32 [1.09,1.60]), CPIS (HR 1.30 [1.08,1.58]) and ACCP definitions (HR 1.22 [1.00,1.47]) were associated with hospital mortality. CONCLUSIONS: Rates of ICU-acquired pneumonia vary by definition and are associated with differential increased risk of death.


Subject(s)
Pneumonia, Ventilator-Associated , Adult , Humans , Cohort Studies , Pneumonia, Ventilator-Associated/microbiology , Intensive Care Units , Hospital Mortality
4.
Crit Care ; 25(1): 436, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34920728

ABSTRACT

BACKGROUND: The measurement of circulating substrate concentrations does not provide information about substrate kinetics. It, therefore, remains unclear if a decrease in plasma concentration of albumin, as seen during critical illness, is a consequence of suppressed production in the liver or increased peripheral clearance. In this study, using stable isotope tracer infusions, we measured albumin and fibrinogen kinetics in septic patients and in a control group of non-septic subjects. METHODS: With the approval from the institutional Research Ethics Board and after obtaining written informed consent from patients or their substitute decision maker, mechanically ventilated patients with sepsis and patients scheduled for elective coronary artery bypass grafting were enrolled. Patients in the non-sepsis group were studied on the day before surgery. The stable isotope L-[ring-2H5]phenylalanine was used to measure absolute synthesis rates (ASR) of albumin and fibrinogen. A priming dose of L-[ring-2H5]phenylalanine (4 µmol/kg) was given followed by a six-hour infusion at a rate of 0.15 µmol/kg/min. At baseline and hourly thereafter, blood was drawn to measure isotope enrichments by gas chromatography/mass spectrometry. Very low density lipoprotein apolipoprotein-B 100 isotopic enrichment was used to represent the isotopic enrichment of the phenylalanine precursor pool from which the liver synthesizes proteins. Plasma albumin and fibrinogen concentrations were also measured. RESULTS: Mean plasma albumin in septic patients was decreased when compared to non-septic patients, while synthesis rates were comparable. Mean plasma fibrinogen and ASR in septic patients was increased when compared to non-septic patients. In non-septic patients, no statistically significant correlation between plasma albumin and ASR was observed but plasma fibrinogen significantly correlated with ASR. In septic patients, plasma albumin and fibrinogen significantly correlated with ASR. CONCLUSIONS: While septic patients showed lower plasma albumin levels than non-septic patients, albumin synthesis was similar in the two groups suggesting that hypoalbuminemia during sepsis was not caused by suppressed hepatic production but a result of enhanced clearance from the circulation. Hyperfibrinogenemia in septic patients was a consequence of increased fibrinogen production. TRIAL REGISTRATION: ClinicalTrials.gov: NCT02865408 (registered on August 12, 2016) and ClinicalTrials.gov: NCT02549443 (registered on September 15, 2015).


Subject(s)
Hypoalbuminemia , Sepsis , Fibrinogen , Humans , Kinetics , Serum Albumin
5.
Crit Care Med ; 49(2): 311-323, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33332817

ABSTRACT

OBJECTIVES: In many jurisdictions, ethical concerns require surrogate humane endpoints to replace death in small animal models of acute lung injury. Heterogenous selection and reporting of surrogate endpoints render interpretation and generalizability of findings between studies difficult. We aimed to establish expert-guided consensus among preclinical scientists and laboratory animal veterinarians on selection and reporting of surrogate endpoints, monitoring of these models, and the use of analgesia. DESIGN: A three-round consensus process, using modified Delphi methodology, with researchers who use small animal models of acute lung injury and laboratory animal veterinarians who provide care for these animals. Statements on the selection and reporting of surrogate endpoints, monitoring, and analgesia were generated through a systematic search of MEDLINE and Embase. Participants were asked to suggest any additional potential statements for evaluation. SETTING: A web-based survey of participants representing the two stakeholder groups (researchers, laboratory animal veterinarians). Statements were rated on level of evidence and strength of support by participants. A final face-to-face meeting was then held to discuss results. SUBJECTS: None. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: Forty-two statements were evaluated, and 29 were rated as important, with varying strength of evidence. The majority of evidence was based on rodent models of acute lung injury. Endpoints with strong support and evidence included temperature changes and body weight loss. Behavioral signs and respiratory distress also received support but were associated with lower levels of evidence. Participants strongly agreed that analgesia affects outcomes in these models and that none may be necessary following nonsurgical induction of acute lung injury. Finally, participants strongly supported transparent reporting of surrogate endpoints. A prototype composite score was also developed based on participant feedback. CONCLUSIONS: We provide a preliminary framework that researchers and animal welfare committees may adapt for their needs. We have identified knowledge gaps that future research should address.


Subject(s)
Acute Lung Injury/physiopathology , Animal Care Committees/organization & administration , Animal Welfare/standards , Animals, Laboratory , Consensus , Animals , Biomarkers , Humans , Models, Animal , Veterinarians/standards
6.
Burns ; 47(4): 765-775, 2021 06.
Article in English | MEDLINE | ID: mdl-33288334

ABSTRACT

INTRODUCTION: A need exists to improve the efficiency of clinical trials in burn care. The objective of this study was to validate "Persistent Organ Dysfunction" plus death as endpoint in burn patients and to demonstrate its statistical efficiency. METHODS: This secondary outcome analysis of a dataset from a prospective international multicenter RCT (RE-ENERGIZE) included patients with burned total body surface area >20% and a 6-month follow-up. Persistent organ dysfunction was defined as persistence of organ dysfunction with life-supportiing technologies and ICU care. RESULTS: In the 539 included patients, the prevalence of 0p p+ pdeath was 40% at day 14 and of 27% at day 28. At both timepoints, survivors with POD (vs. survivors without POD) had a higher mortality rate, longer ICU- and hospital-stays, and a reduced quality of life. POD + death as an endpoint could result in reduced sample size requirements for clinical trials. Detecting a 25% relative risk reduction in 28-day mortality would require a sample size of 4492 patients, whereas 1236 patients would be required were 28-day POD + death used. CONCLUSIONS: POD + death represents a promising composite outcome measure that may reduce the sample size requirements of clinical trials in severe burns patients. Further validation in larger clinical trials is warranted. STUDY TYPE: Prospective cohort study, level of evidence: II.


Subject(s)
Burns/complications , Multiple Organ Failure/etiology , Outcome Assessment, Health Care/standards , Adult , Aged , Burns/epidemiology , Chi-Square Distribution , Female , Humans , Intensive Care Units/organization & administration , Intensive Care Units/statistics & numerical data , Length of Stay/statistics & numerical data , Male , Middle Aged , Multiple Organ Failure/classification , Multiple Organ Failure/epidemiology , Organ Dysfunction Scores , Outcome Assessment, Health Care/trends , Quality of Life/psychology
8.
Crit Care ; 24(1): 499, 2020 08 12.
Article in English | MEDLINE | ID: mdl-32787899

ABSTRACT

The goal of nutrition support is to provide the substrates required to match the bioenergetic needs of the patient and promote the net synthesis of macromolecules required for the preservation of lean mass, organ function, and immunity. Contemporary observational studies have exposed the pervasive undernutrition of critically ill patients and its association with adverse clinical outcomes. The intuitive hypothesis is that optimization of nutrition delivery should improve ICU clinical outcomes. It is therefore surprising that multiple large randomized controlled trials have failed to demonstrate the clinical benefit of restoring or maximizing nutrient intake. This may be in part due to the absence of biological markers that identify patients who are most likely to benefit from nutrition interventions and that monitor the effects of nutrition support. Here, we discuss the need for practical risk stratification tools in critical care nutrition, a proposed rationale for targeted biomarker development, and potential approaches that can be adopted for biomarker identification and validation in the field.


Subject(s)
Biomarkers/analysis , Nutrition Therapy/standards , Albumins/analysis , Biomarkers/blood , Body Composition/physiology , Body Mass Index , C-Reactive Protein/analysis , Critical Care/methods , Critical Care/statistics & numerical data , Enteral Nutrition/adverse effects , Enteral Nutrition/methods , Enteral Nutrition/standards , Humans , Insulin Resistance/physiology , Interleukin-6/analysis , Interleukin-6/blood , Nitrogen/analysis , Nitrogen/blood , Nutrition Therapy/adverse effects , Nutrition Therapy/methods , Nutritional Support/adverse effects , Nutritional Support/methods , Nutritional Support/standards , Parenteral Nutrition/adverse effects , Parenteral Nutrition/methods , Parenteral Nutrition/standards , Proteins/analysis
10.
Sci Rep ; 9(1): 17620, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31772273

ABSTRACT

Inactivation of the protein complex 'mechanistic target of rapamycin complex 1' (mTORC1) can increase the nuclear content of transcriptional regulators of metabolism and apoptosis. Previous studies established that nuclear import of signal transducer and activator of transcription-1 (STAT1) requires the mTORC1-associated adaptor karyopherin-α1 (KPNA1) when mTORC1 activity is reduced. However, the role of other mTORC1-interacting proteins in the complex, including 'protein kinase C delta' (PKCδ), have not been well characterized. In this study, we demonstrate that PKCδ, a STAT1 kinase, contains a functional 'target of rapamycin signaling' (TOS) motif that directs its interaction with mTORC1. Depletion of KPNA1 by RNAi prevented the nuclear import of PKCδ in cells exposed to the mTORC1 inhibitor rapamycin or amino acid restriction. Mutation of the TOS motif in PKCδ led to its loss of regulation by mTORC1 or karyopherin-α1, resulting in increased constitutive nuclear content. In cells expressing wild-type PKCδ, STAT1 activity and apoptosis were increased by rapamycin or interferon-ß. Those expressing the PKCδ TOS mutant exhibited increased STAT1 activity and apoptosis; further enhancement by rapamycin or interferon-ß, however, was lost. Therefore, the TOS motif in PKCδ is a novel structural mechanism by which mTORC1 prevents PKCδ and STAT1 nuclear import, and apoptosis.


Subject(s)
Cell Nucleus/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Protein Kinase C-delta/metabolism , Active Transport, Cell Nucleus/drug effects , Amino Acid Motifs , Apoptosis/drug effects , Apoptosis/physiology , Cell Line , Humans , Models, Molecular , Mutation, Missense , Point Mutation , Protein Conformation , Protein Interaction Mapping , Protein Kinase C-delta/chemistry , Protein Kinase C-delta/genetics , RNA Interference , RNA, Small Interfering/genetics , Recombinant Proteins/metabolism , Regulatory-Associated Protein of mTOR/metabolism , STAT1 Transcription Factor/biosynthesis , Sequence Alignment , Sirolimus/pharmacology , alpha Karyopherins/antagonists & inhibitors , alpha Karyopherins/metabolism
12.
Cell Death Dis ; 9(3): 254, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29449538

ABSTRACT

Oxidative stress determines cell fate through several mechanisms, among which regulation of mRNA translation by the phosphorylation of the alpha (α) subunit of the translation initiation factor eIF2α at serine 51 (eIF2αP) plays a prominent role. Increased eIF2αP can contribute to tumor progression as well as tumor suppression. While eIF2αP is increased in most cells to promote survival and adaptation to different forms of stress, we demonstrate that eIF2αP is reduced in tuberous sclerosis complex 2 (TSC2)-deficient cells subjected to oxidative insults. Decreased eIF2αP in TSC2-deficient cells depends on reactive oxygen species (ROS) production and is associated with a reduced activity of the endoplasmic reticulum (ER)-resident kinase PERK owing to the hyper-activation of the mammalian target of rapamycin complex 1 (mTORC1). Downregulation of PERK activity and eIF2αP is accompanied by increased ROS production and enhanced susceptibility of TSC2-deficient cells to extrinsic pro-oxidant stress. The decreased levels of eIF2αP delay tumor formation of TSC2-deficient cells in immune deficient mice, an effect that is significantly alleviated in mice subjected to an anti-oxidant diet. Our findings reveal a previously unidentified connection between mTORC1 and eIF2αP in TSC2-deficient cells with potential implications in tumor suppression in response to oxidative insults.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Fibroblasts/enzymology , Mechanistic Target of Rapamycin Complex 1/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Tuberous Sclerosis/enzymology , eIF-2 Kinase/metabolism , Animals , Antioxidants/pharmacology , Cell Death , Cells, Cultured , Down-Regulation , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Mice , Mice, SCID , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/prevention & control , Oxidative Stress/drug effects , Phosphorylation , Serine , Signal Transduction , Time Factors , Tuberous Sclerosis/genetics , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 2 Protein/deficiency , Tuberous Sclerosis Complex 2 Protein/genetics , Tumor Burden
13.
Ann Am Thorac Soc ; 14(8): 1239-1247, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28763267

ABSTRACT

Rare respiratory diseases (RRDs) are a heterogeneous group of disorders that collectively represent a significant health care burden. In recent years, strong advocacy and policy initiatives have led to advances in the implementation of research and clinical care for rare diseases. The development of specialized centers and research networks has facilitated support for affected individuals as well as emerging programs in basic, translational, and clinical research. In selected RRDs, subsequent gains in knowledge have informed the development of targeted therapies and effective diagnostic tests, but many gaps persist. There was therefore a desire to identify the elements contributing to an effective translational research program in RRDs. To this end, a workshop was convened in October 2015 with a focus on the implementation of effective transnational research networks and collaborations aimed at developing novel diagnostic and therapeutic tools. Key elements included an emphasis on molecular pathogenesis, the continuing engagement of patient advocacy groups and policy makers, the effective use of preclinical models in the translational research pipeline, and the detailed phenotyping of patient cohorts. During the course of the workshop, current logistical and knowledge gaps were identified, and new solutions or opportunities were highlighted.


Subject(s)
Lung Diseases/diagnosis , Lung Diseases/therapy , Rare Diseases/diagnosis , Rare Diseases/therapy , Translational Research, Biomedical , Animals , Clinical Trials as Topic , Consensus Development Conferences as Topic , Genetic Association Studies , Humans , Lung Diseases/genetics , Mice , Rare Diseases/genetics , Societies, Medical , United States
14.
Cancer Res ; 77(20): 5491-5502, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28830860

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a progressive destructive neoplasm of the lung associated with inactivating mutations in the TSC1 or TSC2 tumor suppressor genes. Cell or animal models that accurately reflect the pathology of LAM have been challenging to develop. Here, we generated a robust human cell model of LAM by reprogramming TSC2 mutation-bearing fibroblasts from a patient with both tuberous sclerosis complex (TSC) and LAM (TSC-LAM) into induced pluripotent stem cells (iPSC), followed by selection of cells that resemble those found in LAM tumors by unbiased in vivo differentiation. We established expandable cell lines under smooth muscle cell (SMC) growth conditions that retained a patient-specific genomic TSC2+/- mutation and recapitulated the molecular and functional characteristics of pulmonary LAM cells. These include multiple indicators of hyperactive mTORC1 signaling, presence of specific neural crest and SMC markers, expression of VEGF-D and female sex hormone receptors, reduced autophagy, and metabolic reprogramming. Intriguingly, the LAM-like features of these cells suggest that haploinsufficiency at the TSC2 locus contributes to LAM pathology, and demonstrated that iPSC reprogramming and SMC lineage differentiation of somatic patient cells with germline mutations was a viable approach to generate LAM-like cells. The patient-derived SMC lines we have developed thus represent a novel cellular model of LAM that can advance our understanding of disease pathogenesis and develop therapeutic strategies against LAM. Cancer Res; 77(20); 5491-502. ©2017 AACR.


Subject(s)
Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/pathology , Myocytes, Smooth Muscle/physiology , Pluripotent Stem Cells/physiology , Animals , Cell Proliferation/physiology , Female , Haploinsufficiency , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology
15.
Nutr Clin Pract ; 32(1_suppl): 58S-71S, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28388372

ABSTRACT

Emerging evidence suggests that exogenous protein/amino acid supplementation has the potential to improve the recovery of critically ill patients. After a careful review of the published evidence, experts have concluded that critically ill patients should receive up to 2.0-2.5 g/kg/d of protein. Despite this, however, recent review of current International Nutrition Survey data suggests that protein in critically ill patients is underprescribed and grossly underdelivered. Furthermore, the survey suggests that most of protein administration comes from enteral nutrition (EN) despite the availability of products and protocols that enhance the delivery of protein/amino acids in the intensive care unit (ICU) setting. While future research clarifies the dose, timing, and composition for exogenous protein administration, as well as identification of patients who will benefit the most, ongoing process improvement initiatives should target a concerted effort to increase protein intake in the critically ill. This assertion follows from the notion that current patients are possibly being harmed while we wait for confirmatory evidence. Further research should also develop better tools to enable bedside practitioners to monitor optimal or adequate protein intake for individual patients. Finally, exploring the effect of combining adequate protein delivery with early mobility and/or resistance exercise in the ICU setting has the greatest potential for improving the functional outcomes of survivors of critical illness and warrants further study.


Subject(s)
Dietary Proteins , Enteral Nutrition/methods , Intensive Care Units , Amino Acids , Critical Illness , Humans , Parenteral Nutrition/methods
16.
J Biol Chem ; 292(5): 1899-1909, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28011640

ABSTRACT

Autophagy involves the lysosomal degradation of cytoplasmic contents for regeneration of anabolic substrates during nutritional or inflammatory stress. Its initiation occurs rapidly after inactivation of the protein kinase mammalian target of rapamycin (mTOR) (or mechanistic target of rapamycin), leading to dephosphorylation of Unc-51-like kinase 1 (ULK1) and autophagosome formation. Recent studies indicate that mTOR can, in parallel, regulate the activity of stress transcription factors, including signal transducer and activator of transcription-1 (STAT1). The current study addresses the role of STAT1 as a transcriptional suppressor of autophagy genes and autophagic activity. We show that STAT1-deficient human fibrosarcoma cells exhibited enhanced autophagic flux as well as its induction by pharmacological inhibition of mTOR. Consistent with enhanced autophagy initiation, ULK1 mRNA and protein levels were increased in STAT1-deficient cells. By chromatin immunoprecipitation, STAT1 bound a putative regulatory sequence in the ULK1 5'-flanking region, the mutation of which increased ULK1 promoter activity, and rendered it unresponsive to mTOR inhibition. Consistent with an anti-apoptotic effect of autophagy, rapamycin-induced apoptosis and cytotoxicity were blocked in STAT1-deficient cells but restored in cells simultaneously exposed to the autophagy inhibitor ammonium chloride. In vivo, skeletal muscle ULK1 mRNA and protein levels as well as autophagic flux were significantly enhanced in STAT1-deficient mice. These results demonstrate a novel mechanism by which STAT1 negatively regulates ULK1 expression and autophagy.


Subject(s)
Autophagy-Related Protein-1 Homolog/biosynthesis , Autophagy/physiology , Gene Expression Regulation, Enzymologic/physiology , Intracellular Signaling Peptides and Proteins/biosynthesis , STAT1 Transcription Factor/metabolism , Animals , Autophagy-Related Protein-1 Homolog/genetics , Cell Line, Tumor , Gene Expression Regulation, Enzymologic/drug effects , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Promoter Regions, Genetic/physiology , STAT1 Transcription Factor/genetics , Sirolimus/pharmacology
17.
Am J Physiol Regul Integr Comp Physiol ; 311(6): R1085-R1092, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27707724

ABSTRACT

Cardiac surgery triggers an inflammatory stress response, leading to protein catabolism, a process that even high-dose insulin therapy alone cannot reverse. To determine whether hyperinsulinemic-normoglycemic clamp and perioperative amino acid (AA) supplementation improves whole body protein balance, 20 patients scheduled for elective coronary artery bypass grafting surgery were randomly assigned to have intra- and postoperative hyperinsulinemic-normoglycemic clamp, with or without intravenous AA supplementation. Primed continuous infusions of [6,6-2H2]glucose and l-[1-13C]leucine were used to quantify whole body protein and glucose metabolism before and after surgery. Adipose tissue and serum cytokines were also analyzed to measure their responsiveness to the anabolic effect of AA administration. During hyperinsulinemic-normoglycemic clamp, AA supplementation successfully stimulated whole body protein synthesis, resulting in a positive whole body protein balance after surgery (insulin: -13.6 ± 4.5 vs. insulin + AA: 2.1 ± 5.4 µmol·kg-1·h-1, P < 0.001). Endogenous glucose production was equally suppressed in both groups (insulin: 0.0 ± 3.8 vs. insulin + AA 1.6 ± 1.6 µmol·kg-1·min-1, P = 0.230). AA supplementation led to significant changes in serum and tissue IL-6 (insulin: 246.6 ± 111.2 vs. insulin + AA: 124.5 ± 79.3 pg/ml, P = 0.011). In conclusion, hyperinsulinemic-normoglycemic clamp technique, together with AA supplementation, can induce an anabolic state after open-heart surgery, as quantified by a positive whole body protein balance.


Subject(s)
Amino Acids/administration & dosage , Coronary Artery Bypass/adverse effects , Inflammation/etiology , Inflammation/metabolism , Insulin/administration & dosage , Protein Biosynthesis/drug effects , Aged , Blood Glucose/drug effects , Blood Glucose/metabolism , Drug Therapy, Combination/methods , Female , Humans , Inflammation/prevention & control , Male , Metabolism/drug effects , Metabolism/physiology , Postoperative Care/methods , Treatment Outcome
18.
Oncotarget ; 7(38): 61152-61165, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27458154

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a destructive lung disease that can arise sporadically or in adults suffering from the tumor syndrome tuberous sclerosis complex (TSC). Microscopic tumors ('LAM nodules') in the lung interstitium arise from lymphatic invasion and metastasis. These consist of smooth muscle-like cells (LAM cells) that exhibit markers of neural crest differentiation and loss of the tumor suppressor protein 'tuberous sclerosis complex-2' (TSC2). Consistent with a neural phenotype, expression of the neuropeptide urotensin-II and its receptor was detected in LAM nodules. We hypothesized that loss of TSC2 sensitizes cells to the oncogenic effects of urotensin-II. TSC2-deficient Eker rat uterine leiomyoma ELT3 cells were stably transfected with empty vector or plasmid for the expression of TSC2. Urotensin-II increased cell viability and proliferation in TSC2-deficient cells, but not in TSC2-reconstituted cells. When exposed to urotensin-II, TSC2-deficient cells exhibited greater migration, anchorage-independent cell growth, and matrix invasion. The effects of urotensin-II on TSC2-deficient cells were blocked by the urotensin receptor antagonist SB657510, and accompanied by activation of Erk mitogen-activated protein kinase and focal adhesion kinase. Urotensin-II-induced proliferation and migration were reproduced in TSC2-deficient human angiomyolipoma cells, but not in those stably expressing TSC2. In a mouse xenograft model, SB657510 blocked the growth of established ELT3 tumors, reduced the number of circulating tumor cells, and attenuated the production of VEGF-D, a clinical biomarker of LAM. Urotensin receptor antagonists may be selective therapeutic agents for the treatment of LAM or other neural crest-derived neoplasms featuring loss of TSC2 or increased expression of the urotensin receptor.


Subject(s)
Tumor Suppressor Proteins/genetics , Urotensins/pharmacology , Uterine Neoplasms/metabolism , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Chemotaxis , Female , Germ-Line Mutation , Humans , Lung Diseases/metabolism , MAP Kinase Signaling System , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplasm Metastasis , Phenotype , Rats , Receptors, G-Protein-Coupled/metabolism , Sulfonamides/pharmacology , Tuberous Sclerosis Complex 2 Protein , Uterine Neoplasms/genetics , Xenograft Model Antitumor Assays
19.
Mol Cancer Res ; 13(10): 1377-88, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26130148

ABSTRACT

UNLABELLED: The mTOR nucleates two complexes, namely mTOR complex 1 and 2 (mTORC1 and mTORC2), which are implicated in cell growth, survival, metabolism, and cancer. Phosphorylation of the α-subunit of translation initiation factor eIF2 at serine 51 (eIF2αS51P) is a key event of mRNA translation initiation and a master regulator of cell fate during cellular stress. Recent studies have implicated mTOR signaling in the stress response, but its connection to eIF2αS51P has remained unclear. Herein, we report that genetic as well as catalytic inhibition of mTORC2 induces eIF2αS51P. On the other hand, the allosteric inhibitor rapamycin induces eIF2αS51P through pathways that are independent of mTORC1 inactivation. Increased eIF2αS51P by impaired mTORC2 depends on the inactivation of AKT, which primes the activation of the endoplasmic reticulum (ER)-resident kinase PERK/PEK. The biologic function of eIF2αS51P was characterized in tuberous sclerosis complex (TSC)-mutant cells, which are defective in mTORC2 and AKT activity. TSC-mutant cells exhibit increased PERK activity, which is downregulated by the reconstitution of the cells with an activated form of AKT1. Also, TSC-mutant cells are increasingly susceptible to ER stress, which is reversed by AKT1 reconstitution. The susceptibility of TSC-mutant cells to ER stress is further enhanced by the pharmacologic inhibition of PERK or genetic inactivation of eIF2αS51P. Thus, the PERK/eIF2αS51P arm is an important compensatory prosurvival mechanism, which substitutes for the loss of AKT under ER stress. IMPLICATIONS: A novel mechanistic link between mTOR function and protein synthesis is identified in TSC-null tumor cells under stress and reveals potential for the development of antitumor treatments with stress-inducing chemotherapeutics.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Fibrosarcoma/drug therapy , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Humans , Mechanistic Target of Rapamycin Complex 2 , Mice , Multiprotein Complexes/antagonists & inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Serine/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Transfection , Up-Regulation , eIF-2 Kinase/metabolism
20.
Biochim Biophys Acta ; 1853(10 Pt A): 2539-52, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26094769

ABSTRACT

Expression and activity of the Ste20-like kinase, SLK, are increased during kidney development and recovery from ischemia-reperfusion injury. SLK mediates apoptosis in various cells, and can regulate cell cycle progression and cytoskeletal remodeling. In cells, SLK is detected in a high molecular mass complex, suggesting that SLK is a dimer/oligomer, or is in tight association with other proteins. To better understand the regulation, localization and function of SLK, we sought to identify proteins in this high molecular mass complex. Analysis by mass spectroscopy identified the nucleoporin, translocated promoter region (Tpr), and the cytoskeletal protein, α-actinin-4, as potential SLK-interacting proteins. Using a protein complementation assay, we showed that the 350 amino acid C-terminal, coiled-coil domain of SLK was responsible for homodimerization, as well as interaction with Tpr and α-actinin-4. The association of SLK with Tpr and α-actinin-4, respectively, was confirmed by co-immunoprecipitation. Subsets of total cellular SLK colocalized with Tpr at the nuclear envelope, and α-actinin-4 in the cytoplasm. Expression of Tpr attenuated activation-specific autophosphorylation of SLK, and blocked SLK-induced apoptosis and AP-1 activity. In contrast to the effect of Tpr, autophosphorylation of SLK was not affected by α-actinin-4. Thus, SLK interacts with Tpr and α-actinin-4 in cells, and these protein-protein interactions may control the subcellular localization and the biological activity of SLK.


Subject(s)
Actinin/metabolism , Nuclear Pore Complex Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Actinin/genetics , Animals , COS Cells , Chlorocebus aethiops , Humans , Nuclear Pore Complex Proteins/genetics , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Protein Transport/physiology , Proto-Oncogene Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...