Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Ultrasound Med ; 43(6): 1121-1129, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38421056

ABSTRACT

OBJECTIVES: We sought to determine the association between intrauterine device (IUD) malposition and previous cesarean delivery (CD) and related uterine anatomical changes. METHODS: A retrospective cohort of all persons with an IUD presenting for two- and three-dimensional pelvic ultrasonography over 2 years, for any gynecologic indication, was compiled. IUD malposition was defined as IUD partially or completely positioned outside the endometrial cavity. Uterine position, uterine flexion, and cesarean scar defect (CSD) size were assessed. Patient characteristics and sonographic findings were compared between those with normally positioned and malpositioned IUD. Primary outcome was the rate of IUD malposition in persons with and without a history of CD. Logistic regression analysis was used to control for potential confounders. RESULTS: Two hundred ninety-six persons with an IUD had a pelvic ultrasound, 240 (81.1%) had a normally positioned IUD, and 56 (18.9%) had a malpositioned IUD. The most common location of IUD malposition was low uterine segment and cervix (67.9%). Malpositioned IUD was associated with referral for evaluation of pelvic pain (P = .001). Prior CD was significantly associated with a malpositioned IUD, after adjusting for confounders (aOR 3.50, 95% CI 1.31-9.35, P = .01). Among persons with prior CD, uterine retroflexion and a large CSD were independent risk factors for IUD malposition (aOR 4.1, 95% CI 1.1-15.9, P = .04 and aOR 5.4, 95% CI 1.4-20.9, P = .01, respectively). CONCLUSIONS: Prior CD is associated with significantly increased risk of IUD malposition. Among persons with previous CD, those with a retroflexed uterus and a large CSD are more likely to have a malpositioned IUD.


Subject(s)
Cesarean Section , Intrauterine Devices , Ultrasonography , Uterus , Humans , Female , Uterus/diagnostic imaging , Retrospective Studies , Adult , Cesarean Section/adverse effects , Ultrasonography/methods , Intrauterine Devices/adverse effects , Cohort Studies , Middle Aged , Imaging, Three-Dimensional/methods , Pregnancy
2.
Cell Rep ; 42(12): 113523, 2023 12 26.
Article in English | MEDLINE | ID: mdl-38060446

ABSTRACT

Ubiquitination of proliferating cell nuclear antigen (PCNA) at lysine 164 (K164) activates DNA damage tolerance pathways. Currently, we lack a comprehensive understanding of how PCNA K164 ubiquitination promotes genome stability. To evaluate this, we generated stable cell lines expressing PCNAK164R from the endogenous PCNA locus. Our data reveal that the inability to ubiquitinate K164 causes perturbations in global DNA replication. Persistent replication stress generates under-replicated regions and is exacerbated by the DNA polymerase inhibitor aphidicolin. We show that these phenotypes are due, in part, to impaired Fanconi anemia group D2 protein (FANCD2)-dependent mitotic DNA synthesis (MiDAS) in PCNAK164R cells. FANCD2 mono-ubiquitination is significantly reduced in PCNAK164R mutants, leading to reduced chromatin association and foci formation, both prerequisites for FANCD2-dependent MiDAS. Furthermore, K164 ubiquitination coordinates direct PCNA/FANCD2 colocalization in mitotic nuclei. Here, we show that PCNA K164 ubiquitination maintains human genome stability by promoting FANCD2-dependent MiDAS to prevent the accumulation of under-replicated DNA.


Subject(s)
DNA Repair , Fanconi Anemia Complementation Group D2 Protein , Humans , DNA/metabolism , DNA Damage , DNA Replication , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Genomic Instability , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitination
3.
Genes Dev ; 37(19-20): 929-943, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37932012

ABSTRACT

The mismatch repair (MMR) deficiency of cancer cells drives mutagenesis and offers a useful biomarker for immunotherapy. However, many MMR-deficient (MMR-d) tumors do not respond to immunotherapy, highlighting the need for alternative approaches to target MMR-d cancer cells. Here, we show that inhibition of the ATR kinase preferentially kills MMR-d cancer cells. Mechanistically, ATR inhibitor (ATRi) imposes synthetic lethality on MMR-d cells by inducing DNA damage in a replication- and MUS81 nuclease-dependent manner. The DNA damage induced by ATRi is colocalized with both MSH2 and PCNA, suggesting that it arises from DNA structures recognized by MMR proteins during replication. In syngeneic mouse models, ATRi effectively reduces the growth of MMR-d tumors. Interestingly, the antitumor effects of ATRi are partially due to CD8+ T cells. In MMR-d cells, ATRi stimulates the accumulation of nascent DNA fragments in the cytoplasm, activating the cGAS-mediated interferon response. The combination of ATRi and anti-PD-1 antibody reduces the growth of MMR-d tumors more efficiently than ATRi or anti-PD-1 alone, showing the ability of ATRi to augment the immunotherapy of MMR-d tumors. Thus, ATRi selectively targets MMR-d tumor cells by inducing synthetic lethality and enhancing antitumor immunity, providing a promising strategy to complement and augment MMR deficiency-guided immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , DNA Mismatch Repair , Animals , Mice , DNA Mismatch Repair/genetics , Synthetic Lethal Mutations , DNA , Immunotherapy
4.
Cell Rep ; 42(7): 112792, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37454295

ABSTRACT

The ATR kinase safeguards genomic integrity during S phase, but how ATR protects DNA replication forks remains incompletely understood. Here, we combine four distinct assays to analyze ATR functions at ongoing and newly assembled replication forks upon replication inhibition by hydroxyurea. At ongoing forks, ATR inhibitor (ATRi) increases MRE11- and EXO1-mediated nascent DNA degradation from PrimPol-generated, single-stranded DNA (ssDNA) gaps. ATRi also exposes template ssDNA through fork uncoupling and nascent DNA degradation. Electron microscopy reveals that ATRi reduces reversed forks by increasing gap-dependent nascent DNA degradation. At new forks, ATRi triggers MRE11- and CtIP-initiated template DNA degradation by EXO1, exposing nascent ssDNA. Upon PARP inhibition, ATRi preferentially exacerbates gap-dependent nascent DNA degradation at ongoing forks in BRCA1/2-deficient cells and disrupts the restored gap protection in BRCA1-deficient, PARP-inhibitor-resistant cells. Thus, ATR protects ongoing and new forks through distinct mechanisms, providing an extended view of ATR's functions in stabilizing replication forks.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , BRCA1 Protein , DNA-Binding Proteins , BRCA1 Protein/metabolism , BRCA2 Protein/metabolism , DNA Replication , DNA, Single-Stranded , DNA-Binding Proteins/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Humans , Ataxia Telangiectasia Mutated Proteins/metabolism
5.
Mol Cell ; 82(21): 3985-4000.e4, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36265486

ABSTRACT

Alternative lengthening of telomeres (ALT), a telomerase-independent process maintaining telomeres, is mediated by break-induced replication (BIR). RAD52 promotes ALT by facilitating D-loop formation, but ALT also occurs through a RAD52-independent BIR pathway. Here, we show that the telomere non-coding RNA TERRA forms dynamic telomeric R-loops and contributes to ALT activity in RAD52 knockout cells. TERRA forms R-loops in vitro and at telomeres in a RAD51AP1-dependent manner. The formation of R-loops by TERRA increases G-quadruplexes (G4s) at telomeres. G4 stabilization enhances ALT even when TERRA is depleted, suggesting that G4s act downstream of R-loops to promote BIR. In vitro, the telomeric R-loops assembled by TERRA and RAD51AP1 generate G4s, which persist after R-loop resolution and allow formation of telomeric D-loops without RAD52. Thus, the dynamic telomeric R-loops formed by TERRA and RAD51AP1 enable the RAD52-independent ALT pathway, and G4s orchestrate an R- to D-loop switch at telomeres to stimulate BIR.


Subject(s)
RNA, Long Noncoding , Telomerase , Telomere Homeostasis , Telomere/genetics , Telomere/metabolism , Telomerase/genetics , Telomerase/metabolism , R-Loop Structures/genetics , DNA Repair
6.
Blood ; 138(2): 190-198, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33895804

ABSTRACT

Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is associated with thrombotic complications in adults, but the incidence of COVID-19-related thrombosis in children and adolescents is unclear. Most children with acute COVID-19 have mild disease, but coagulopathy has been associated with multisystem inflammatory syndrome in children (MIS-C), a postinfectious complication. We conducted a multicenter retrospective cohort study to determine the incidence of thrombosis in children hospitalized with COVID-19 or MIS-C and evaluate associated risk factors. We classified patients into 1 of 3 groups for analysis: COVID-19, MIS-C, or asymptomatic SARS-CoV-2. Among a total of 853 admissions (COVID-19, n = 426; MIS-C, n = 138; and asymptomatic SARS-CoV-2, n = 289) in 814 patients, there were 20 patients with thrombotic events (TEs; including 1 stroke). Patients with MIS-C had the highest incidence (9 [6.5%] of 138) vs COVID-19 (9 [2.1%] of 426) or asymptomatic SARS-CoV-2 (2 [0.7%] of 289). In patients with COVID-19 or MIS-C, a majority of TEs (89%) occurred in patients age ≥12 years. Patients age ≥12 years with MIS-C had the highest rate of thrombosis at 19% (9 of 48). Notably, 71% of TEs that were not present on admission occurred despite thromboprophylaxis. Multivariable analysis identified the following as significantly associated with thrombosis: age ≥12 years, cancer, presence of a central venous catheter, and MIS-C. In patients with COVID-19 or MIS-C, hospital mortality was 2.3% (13 of 564), but it was 28% (5 of 18) in patients with TEs. Our findings may help inform pediatric thromboprophylaxis strategies.


Subject(s)
COVID-19/complications , Systemic Inflammatory Response Syndrome/complications , Thrombosis/etiology , Adolescent , Adult , Age Factors , Anticoagulants/therapeutic use , COVID-19/diagnosis , Child , Child, Preschool , Female , Humans , Infant , Male , Retrospective Studies , Risk Factors , SARS-CoV-2/isolation & purification , Systemic Inflammatory Response Syndrome/diagnosis , Thrombosis/drug therapy , Thrombosis/prevention & control , Young Adult
7.
Nat Commun ; 12(1): 1626, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712616

ABSTRACT

Minichromosome maintenance protein 10 (MCM10) is essential for eukaryotic DNA replication. Here, we describe compound heterozygous MCM10 variants in patients with distinctive, but overlapping, clinical phenotypes: natural killer (NK) cell deficiency (NKD) and restrictive cardiomyopathy (RCM) with hypoplasia of the spleen and thymus. To understand the mechanism of MCM10-associated disease, we modeled these variants in human cell lines. MCM10 deficiency causes chronic replication stress that reduces cell viability due to increased genomic instability and telomere erosion. Our data suggest that loss of MCM10 function constrains telomerase activity by accumulating abnormal replication fork structures enriched with single-stranded DNA. Terminally-arrested replication forks in MCM10-deficient cells require endonucleolytic processing by MUS81, as MCM10:MUS81 double mutants display decreased viability and accelerated telomere shortening. We propose that these bi-allelic variants in MCM10 predispose specific cardiac and immune cell lineages to prematurely arrest during differentiation, causing the clinical phenotypes observed in both NKD and RCM patients.


Subject(s)
Alleles , Cardiomyopathies/genetics , Minichromosome Maintenance Proteins/genetics , Minichromosome Maintenance Proteins/immunology , Telomere Shortening , Cell Cycle Proteins/metabolism , Cell Line , DNA Replication , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endonucleases/genetics , Endonucleases/metabolism , Humans , Killer Cells, Natural
8.
Clin Infect Dis ; 72(1): 82-89, 2021 01 23.
Article in English | MEDLINE | ID: mdl-32634822

ABSTRACT

BACKGROUND: Understanding nosocomial acquisition, outbreaks, and transmission chains in real time will be fundamental to ensuring infection-prevention measures are effective in controlling coronavirus disease 2019 (COVID-19) in healthcare. We report the design and implementation of a hospital-onset COVID-19 infection (HOCI) surveillance system for an acute healthcare setting to target prevention interventions. METHODS: The study took place in a large teaching hospital group in London, United Kingdom. All patients tested for SARS-CoV-2 between 4 March and 14 April 2020 were included. Utilizing data routinely collected through electronic healthcare systems we developed a novel surveillance system for determining and reporting HOCI incidence and providing real-time network analysis. We provided daily reports on incidence and trends over time to support HOCI investigation and generated geotemporal reports using network analysis to interrogate admission pathways for common epidemiological links to infer transmission chains. By working with stakeholders the reports were co-designed for end users. RESULTS: Real-time surveillance reports revealed changing rates of HOCI throughout the course of the COVID-19 epidemic, key wards fueling probable transmission events, HOCIs overrepresented in particular specialties managing high-risk patients, the importance of integrating analysis of individual prior pathways, and the value of co-design in producing data visualization. Our surveillance system can effectively support national surveillance. CONCLUSIONS: Through early analysis of the novel surveillance system we have provided a description of HOCI rates and trends over time using real-time shifting denominator data. We demonstrate the importance of including the analysis of patient pathways and networks in characterizing risk of transmission and targeting infection-control interventions.


Subject(s)
COVID-19 , Hospitals , Humans , London , SARS-CoV-2 , United Kingdom
9.
Nat Commun ; 11(1): 2147, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32358495

ABSTRACT

Upon genotoxic stress, PCNA ubiquitination allows for replication of damaged DNA by recruiting lesion-bypass DNA polymerases. However, PCNA is also ubiquitinated during normal S-phase progression. By employing 293T and RPE1 cells deficient in PCNA ubiquitination, generated through CRISPR/Cas9 gene editing, here, we show that this modification promotes cellular proliferation and suppression of genomic instability under normal growth conditions. Loss of PCNA-ubiquitination results in DNA2-dependent but MRE11-independent nucleolytic degradation of nascent DNA at stalled replication forks. This degradation is linked to defective gap-filling in the wake of the replication fork and incomplete Okazaki fragment maturation, which interferes with efficient PCNA unloading by ATAD5 and subsequent nucleosome deposition by CAF-1. Moreover, concomitant loss of PCNA-ubiquitination and the BRCA pathway results in increased nascent DNA degradation and PARP inhibitor sensitivity. In conclusion, we show that by ensuring efficient Okazaki fragment maturation, PCNA-ubiquitination protects fork integrity and promotes the resistance of BRCA-deficient cells to PARP-inhibitors.


Subject(s)
Proliferating Cell Nuclear Antigen/metabolism , Cell Line, Tumor , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , Comet Assay , DNA/genetics , DNA Damage/genetics , DNA Damage/physiology , DNA Repair/genetics , DNA Repair/physiology , DNA Replication/genetics , DNA Replication/physiology , Fluorescent Antibody Technique , Genomic Instability/genetics , Genomic Instability/physiology , HEK293 Cells , HeLa Cells , Humans , Proliferating Cell Nuclear Antigen/genetics , Protein Binding , Ubiquitination/genetics , Ubiquitination/physiology
10.
Genes (Basel) ; 10(1)2018 Dec 24.
Article in English | MEDLINE | ID: mdl-30586904

ABSTRACT

DNA damage is a constant source of stress challenging genomic integrity. To ensure faithful duplication of our genomes, mechanisms have evolved to deal with damage encountered during replication. One such mechanism is referred to as DNA damage tolerance (DDT). DDT allows for replication to continue in the presence of a DNA lesion by promoting damage bypass. Two major DDT pathways exist: error-prone translesion synthesis (TLS) and error-free template switching (TS). TLS recruits low-fidelity DNA polymerases to directly replicate across the damaged template, whereas TS uses the nascent sister chromatid as a template for bypass. Both pathways must be tightly controlled to prevent the accumulation of mutations that can occur from the dysregulation of DDT proteins. A key regulator of error-prone versus error-free DDT is the replication clamp, proliferating cell nuclear antigen (PCNA). Post-translational modifications (PTMs) of PCNA, mainly by ubiquitin and SUMO (small ubiquitin-like modifier), play a critical role in DDT. In this review, we will discuss the different types of PTMs of PCNA and how they regulate DDT in response to replication stress. We will also cover the roles of PCNA PTMs in lagging strand synthesis, meiotic recombination, as well as somatic hypermutation and class switch recombination.

11.
Transfus Med Rev ; 32(4): 244-248, 2018 10.
Article in English | MEDLINE | ID: mdl-30025661

ABSTRACT

The increasing incidence of placenta accreta has paralleled the rise in its greatest risk factor: cesarean delivery. In placenta accreta, the abnormal invasion of the chorionic villi into the myometrium prevents separation of the placenta at delivery, and the myometrium is unable to contract to prevent hemorrhage. Spontaneous uterine rupture and hemoperitoneum may also occur in the setting of placenta percreta. The average blood loss during a delivery complicated by placenta accreta is 2 to 5 L, compared to less than 0.5 L for a normal spontaneous vaginal delivery and less than 1 L for a cesarean delivery. Transfusion support for these patients, including preoperative blood component planning, is challenging for the transfusion service, and there is no consensus on how transfusion services should prepare for such cases. Herein, we review the value of a multidisciplinary approach in minimizing and supporting maternal hemorrhage in placenta accreta, predictors of hemorrhage, blood product preparation, potential strategies to limit blood loss, and intraoperative management considerations. We also highlight future opportunities and challenges in this unique group of patients.


Subject(s)
Blood Transfusion/methods , Placenta Accreta/therapy , Postpartum Hemorrhage/prevention & control , Transfusion Medicine/methods , Antifibrinolytic Agents/therapeutic use , Blood Component Transfusion , Cesarean Section , Female , Hemorrhage , Humans , Interdisciplinary Communication , Placenta/pathology , Postpartum Period , Pregnancy , Preoperative Period , Retrospective Studies
12.
Nucleic Acids Res ; 46(11): 5634-5650, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29741650

ABSTRACT

Overexpression of the flap endonuclease FEN1 has been observed in a variety of cancer types and is a marker for poor prognosis. To better understand the cellular consequences of FEN1 overexpression we utilized a model of its Saccharomyces cerevisiae homolog, RAD27. In this system, we discovered that flap endonuclease overexpression impedes replication fork progression and leads to an accumulation of cells in mid-S phase. This was accompanied by increased phosphorylation of the checkpoint kinase Rad53 and histone H2A-S129. RAD27 overexpressing cells were hypersensitive to treatment with DNA damaging agents, and defective in ubiquitinating the replication clamp proliferating cell nuclear antigen (PCNA) at lysine 164. These effects were reversed when the interaction between overexpressed Rad27 and PCNA was ablated, suggesting that the observed phenotypes were linked to problems in DNA replication. RAD27 overexpressing cells also exhibited an unexpected dependence on the SUMO ligases SIZ1 and MMS21 for viability. Importantly, we found that overexpression of FEN1 in human cells also led to phosphorylation of CHK1, CHK2, RPA32 and histone H2AX, all markers of genome instability. Our data indicate that flap endonuclease overexpression is a driver of genome instability in yeast and human cells that impairs DNA replication in a manner dependent on its interaction with PCNA.


Subject(s)
DNA Damage , Flap Endonucleases/metabolism , Genomic Instability , Proliferating Cell Nuclear Antigen/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , Lung Neoplasms/enzymology , Small Cell Lung Carcinoma/enzymology , Sumoylation , Ubiquitination
13.
Sports Biomech ; 16(2): 238-247, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27593384

ABSTRACT

Compared to competitive runners, recreational runners appear to be more prone to injuries, which have been associated with foot strike patterns. Surprisingly, only few studies had examined the foot strike patterns outside laboratories. Therefore, this study compared the foot strike patterns in recreational runners at outdoor tracks with previously reported data. We also investigated the relationship between foot strike pattern, speed, and footwear in this cohort. Among 434 recreational runners analysed, 89.6% of them landed with rearfoot strike (RFS). Only 6.9 and 3.5% landed with midfoot and forefoot, respectively. A significant shift towards non-RFS was observed in our cohort, when compared with previously reported data. When speed increased by 1 m/s, the odds of having forefoot strike and midfoot strike relative to RFS increased by 2.3 times and 2.6 times, respectively. Runners were 9.2 times more likely to run with a forefoot strike in minimalists compared to regular running shoes, although 70% of runners in minimalists continued to use a RFS. These findings suggest that foot strike pattern may differ across running conditions and runners should consider these factors in order to mitigate potential injury.


Subject(s)
Foot/physiology , Gait/physiology , Running/physiology , Shoes , Adult , Athletic Injuries/prevention & control , Biomechanical Phenomena , Equipment Design , Female , Humans , Male , Running/injuries
14.
FEBS J ; 283(23): 4244-4246, 2016 12.
Article in English | MEDLINE | ID: mdl-27921370

ABSTRACT

The processing of Okazaki fragments when they are assembled into nucleosomes has received little attention. In this issue of The FEBS Journal, Seo and colleagues show that binding to histone tails stimulates the enzymatic activity of flap endonuclease 1 (Rad27). Histone tails are structurally similar to the C terminus of Rad27 and can thus mimic its autostimulatory function. This study highlights an active regulatory role for nucleosomes on DNA metabolism.


Subject(s)
Histones/genetics , Saccharomyces cerevisiae Proteins/genetics , DNA/genetics , DNA Replication , Enzyme Activation , Flap Endonucleases/genetics , Nucleosomes , Saccharomyces cerevisiae/enzymology
15.
J Neurochem ; 136(2): 250-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26438054

ABSTRACT

Neuregulin-1 (NRG1) is an endogenous growth factor with multiple functions in the embryonic and postnatal brain. The NRG1 gene is large and complex, transcribing more than twenty transmembrane proteins and generating a large number of isoforms in tissue and cell type-specific patterns. Within the brain, NRG1 functions have been studied most extensively in neurons and glia, as well as in the peripheral vasculature. Recently, NRG1 signaling has been found to be important in the function of brain microvascular endothelial cells, decreasing IL-1ß-induced increases in endothelial permeability. In the current experiments, we have investigated the pathways through which the NRG1-ß isoform acts on IL-1ß-induced endothelial permeability. Our data show that NRG1-ß increases barrier function, measured by transendothelial electrical resistance, and decreases IL-1ß-induced hyperpermeability, measured by dextran-40 extravasation through a monolayer of brain microvascular endothelial cells plated on transwells. An investigation of key signaling proteins suggests that the effect of NRG1-ß on endothelial permeability is mediated through RhoA activation and myosin light chain phosphorylation, events which affect filamentous actin morphology. In addition, AG825, an inhibitor of the erbB2-associated tyrosine kinase, reduces the effect of NRG1-ß on IL-1ß-induced RhoA activation and myosin light chain phosphorylation. These data add to the evidence that NRG1-ß signaling affects changes in the brain microvasculature in the setting of neuroinflammation. We propose the following events for neuregulin-1-mediated effects on Interleukin-1 ß (IL-1ß)-induced endothelial hyperpermeability: IL-1ß leads to RhoA activation, resulting in an increase in phosphorylation of myosin light chain (MLC). Phosphorylation of MLC is known to result in actin contraction and alterations in the f-actin cytoskeletal structure. These changes are associated with increased endothelial permeability. Neuregulin-1ß acts through its transmembrane receptors to activate intracellular signaling pathways which inhibit IL-1ß-induced RhoA activation and MLC phosphorylation, thereby preserving the f-actin cytoskeletal structure and endothelial barrier function.


Subject(s)
Endothelial Cells/drug effects , Interleukin-1beta/pharmacology , Myosin Light Chains/metabolism , Neuregulin-1/pharmacology , Signal Transduction/physiology , rho-Associated Kinases/metabolism , Adolescent , Benzothiazoles/pharmacology , Brain , Cells, Cultured , Electric Impedance , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Male , Middle Aged , Neuregulin-1/metabolism , Permeability/drug effects , Phosphorylation/drug effects , Signal Transduction/drug effects , Tyrphostins/pharmacology , Young Adult
16.
J Neurotrauma ; 33(14): 1303-16, 2016 07 15.
Article in English | MEDLINE | ID: mdl-26528928

ABSTRACT

Perturbations in blood vessels play a critical role in the pathophysiology of brain injury and neurodegeneration. Here, we use a systematic genome-wide transcriptome screening approach to investigate the vasculome after brain trauma in mice. Mice were subjected to controlled cortical impact and brains were extracted for analysis at 24 h post-injury. The core of the traumatic lesion was removed and then cortical microvesels were isolated from nondirectly damaged ipsilateral cortex. Compared to contralateral cortex and normal cortex from sham-operated mice, we identified a wide spectrum of responses in the vasculome after trauma. Up-regulated pathways included those involved in regulation of inflammation and extracellular matrix processes. Decreased pathways included those involved in regulation of metabolism, mitochondrial function, and transport systems. These findings suggest that microvascular perturbations can be widespread and not necessarily localized to core areas of direct injury per se and may further provide a broader gene network context for existing knowledge regarding inflammation, metabolism, and blood-brain barrier alterations after brain trauma. Further efforts are warranted to map the vasculome with higher spatial and temporal resolution from acute to delayed phase post-trauma. Investigating the widespread network responses in the vasculome may reveal potential mechanisms, therapeutic targets, and biomarkers for traumatic brain injury.


Subject(s)
Brain Injuries, Traumatic , Brain/blood supply , Cerebral Cortex/injuries , Microvessels , Transcriptome , Animals , Brain/immunology , Brain/metabolism , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Microvessels/immunology , Microvessels/metabolism
17.
Transl Stroke Res ; 6(2): 116-24, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24863743

ABSTRACT

Neuroinflammation contributes to the pathophysiology of diverse diseases including stroke, traumatic brain injury, Alzheimer's disease, Parkinson's disease, and multiple sclerosis, resulting in neurodegeneration and loss of neurological function. The response of the microvascular endothelium often contributes to neuroinflammation. One such response is the upregulation of endothelial adhesion molecules which facilitate neutrophil adhesion to the endothelium and their migration from blood to tissue. Neuregulin-1 (NRG1) is an endogenous growth factor which has been reported to have anti-inflammatory effects in experimental stroke models. We hypothesized that NRG1 would decrease the endothelial response to inflammation and result in a decrease in neutrophil adhesion to endothelial cells. We tested this hypothesis in an in vitro model of cytokine-induced endothelial injury, in which human brain microvascular endothelial cells (BMECs) were treated with IL-1ß, along with co-incubation with vehicle or NRG1-ß. Outcome measures included protein levels of endothelial ICAM-1, VCAM-1, and E-selectin, as well as the number of neutrophils that adhere to the endothelial monolayer. Our data show that NRG1-ß decreased the levels of VCAM-1, E-selectin, and neutrophil adhesion to brain microvascular endothelial cells activated by IL1-ß. These findings open new possibilities for investigating NRG1 in neuroprotective strategies in brain injury.


Subject(s)
Cell Adhesion/drug effects , Endothelial Cells/drug effects , Endothelium, Vascular/cytology , Interleukin-1beta/pharmacology , Neuregulin-1/pharmacology , Neutrophils/drug effects , Analysis of Variance , Animals , Brain/anatomy & histology , Cells, Cultured , Dose-Response Relationship, Drug , E-Selectin/metabolism , Female , Humans , Intercellular Adhesion Molecule-1/metabolism , L-Lactate Dehydrogenase/metabolism , Mice , Neutrophils/physiology , Time Factors , Vascular Cell Adhesion Molecule-1/metabolism
18.
Stroke ; 45(7): 2085-92, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24916903

ABSTRACT

BACKGROUND AND PURPOSE: We explored the hypothesis that injured neurons release lipocalin-2 as a help me signal. METHODS: In vivo lipocalin-2 responses were assessed in rat focal cerebral ischemia and human stroke brain samples using a combination of ELISA and immunostaining. In vitro, microglia and astrocytes were exposed to lipocalin-2, and various markers and assays of glial activation were quantified. Functional relevance of neuron-to-glia lipocalin-2 signaling was examined by transferring conditioned media from lipocalin-2-activated microglia and astrocytes onto neurons to see whether activated glia could protect neurons against oxygen-glucose deprivation and promote neuroplasticity. RESULTS: In human stroke samples and rat cerebral ischemia, neuronal expression of lipocalin-2 was significantly increased. In primary cell cultures, exposing microglia and astrocytes to lipocalin-2 resulted in glial activation. In microglia, lipocalin-2 converted resting ramified shapes into a long-rod morphology with reduced branching, increased interleukin-10 release, and enhanced phagocytosis. In astrocytes, lipocalin-2 upregulated glial fibrillary acid protein, brain-derived neurotropic factor, and thrombospondin-1. Conditioned media from lipocalin-2-treated astrocytes upregulated synaptotagmin, and conditioned media from lipocalin-2-treated microglia upregulated synaptophysin and post-synaptic density 95 (PSD95) and protected neurons against oxygen-glucose deprivation. CONCLUSIONS: These findings provide proof of concept that lipocalin-2 is released by injured neurons as a help me distress signal that activates microglia and astrocytes into potentially prorecovery phenotypes.


Subject(s)
Acute-Phase Proteins/metabolism , Astrocytes/metabolism , Brain Ischemia/metabolism , Cerebral Cortex/metabolism , Lipocalins/metabolism , Microglia/metabolism , Neurons/metabolism , Proto-Oncogene Proteins/metabolism , Stroke/metabolism , Aged , Animals , Biomarkers/metabolism , Cerebral Cortex/pathology , Female , Humans , Lipocalin-2 , Male , Neuronal Plasticity/physiology , Phenotype , Rats , Rats, Wistar , Single-Blind Method
19.
J Cereb Blood Flow Metab ; 34(5): 870-5, 2014 May.
Article in English | MEDLINE | ID: mdl-24549187

ABSTRACT

To date, only limited data are available on the effects of pretreatment with novel oral anticoagulants in the event of traumatic brain injury (TBI). We determined intracerebral hemorrhage volume and functional outcome in a standardized TBI model in mice treated with warfarin or dabigatran. Additionally, we investigated whether excess concentrations of dabigatran could increase bleeding and whether this was preventable by using prothrombin complex concentrate (PCC). C57 mice were treated orally with warfarin or dabigatran; sham-treated mice served as controls. Effective anticoagulation was verified by measurement of international normalized ratio and diluted thrombin time, and TBI was induced by controlled cortical impact (CCI). Twenty-four hours after CCI, intracerebral hemorrhage volume was larger in warfarin-pretreated mice than in controls (10.1 ± 4.9 vs 4.1 ± 1.7 µL; analysis of variance post hoc P=0.001), but no difference was found between controls and dabigatran-pretreated mice (5.3 ± 1.5 µL). PCC applied 30 minutes after CCI did not reliably reduce intracerebral hemorrhage induced by excess dabigatran concentration compared with saline (10.4 ± 11.2 vs 8.7 ± 7.1 µL). Our data suggest pathophysiological differences in TBI occurring during warfarin and dabigatran anticoagulation. The reduced hemorrhage formation under dabigatran therapy could present a safety advantage compared with warfarin. An excess dabigatran concentration, however, can increase hemorrhage.


Subject(s)
Benzimidazoles/therapeutic use , Brain Injuries/blood , Brain Injuries/drug therapy , Warfarin/therapeutic use , beta-Alanine/analogs & derivatives , Animals , Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Anticoagulants/therapeutic use , Benzimidazoles/administration & dosage , Benzimidazoles/adverse effects , Blood Coagulation/drug effects , Blood Coagulation Factors/therapeutic use , Brain/drug effects , Brain/pathology , Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/prevention & control , Dabigatran , Hemorrhage/chemically induced , Mice , Mice, Inbred C57BL , Warfarin/administration & dosage , Warfarin/adverse effects , beta-Alanine/administration & dosage , beta-Alanine/adverse effects , beta-Alanine/therapeutic use
20.
Methods Mol Biol ; 1135: 393-402, 2014.
Article in English | MEDLINE | ID: mdl-24510881

ABSTRACT

Common methods for studying angiogenesis in vitro include the tube formation assay, the migration assay, and the study of the endothelial genome. The formation of capillary-like tubes in vitro on basement membrane matrix mimics many steps of the angiogenesis process in vivo and is used widely as a screening test for angiogenic or antiangiogenic factors. Other assays related to the study of angiogenesis include the cell migration assay, the study of gene expression changes during the process of angiogenesis, and the study of endothelial-derived microparticles. Protocols for these procedures will be described here.


Subject(s)
Cell Movement , Endothelial Cells/physiology , Animals , Cell Culture Techniques , Cell Line , Cell Separation , Cell-Derived Microparticles/physiology , Endothelium, Vascular/cytology , Gene Expression Profiling , Mice , Microvessels/cytology , Neovascularization, Physiologic , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , RNA, Messenger/metabolism , Rats , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...