66 research outputs found

    An automated Fpg-based FADU method for the detection of oxidative DNA lesions and screening of antioxidants

    Get PDF
    The oxidation of guanine to 8-oxo-2′-deoxyguanosine (8-oxo-dG) is one of the most abundant and best studied oxidative DNA lesions and is commonly used as a biomarker for oxidative stress. Over the last decades, various methods for the detection of DNA oxidation products have been established and optimized. However, some of them lack sensitivity or are prone to artifact formation, while others are time-consuming, which hampers their application in screening approaches. In this study, we present a formamidopyrimidine glycosylase (Fpg)-based method to detect oxidative lesions in isolated DNA using a modified protocol of the automated version of the fluorimetric detection of alkaline DNA unwinding (FADU) method, initially developed for the measurement of DNA strand breaks (Moreno-Villanueva et al., 2009. BMC Biotechnol. 9, 39). The FADU-Fpg method was validated using a plasmid DNA model, mimicking mitochondrial DNA, and the results were correlated to 8-oxo-dG levels as measured by LC–MS/MS. The FADU-Fpg method can be applied to analyze the potential of compounds to induce DNA strand breaks and oxidative lesions, as exemplified here by treating plasmid DNA with the peroxynitrite-generating molecule Sin-1. Moreover, this method can be used to screen DNA-protective effects of antioxidant substances, as exemplified here for a small-molecule, i.e., uric acid, and a protein, i.e., manganese superoxide dismutase, both of which displayed a dose-dependent protection against the generation of oxidative DNA lesions. In conclusion, the automated FADU-Fpg method offers a rapid and reliable measurement for the detection of peroxynitrite-mediated DNA damage in a cell-free system, rendering it an ideal method for screening the DNA-protective effects of antioxidant compounds.Deutsche Forschungsgemeinschaft (Grant BU 698/6-1)National Institutes of Health (U.S.) (Grant ES002109)National Institutes of Health (U.S.) (Grant CA026731

    Cell-Free DNA and Active Rejection in Kidney Allografts

    Get PDF
    Histologic analysis of the allograft biopsy specimen is the standard method used to differentiate rejection from other injury in kidney transplants. Donor-derived cell-free DNA (dd-cfDNA) is a noninvasive test of allograft injury that may enable more frequent, quantitative, and safer assessment of allograft rejection and injury status. To investigate this possibility, we prospectively collected blood specimens at scheduled intervals and at the time of clinically indicated biopsies. In 102 kidney recipients, we measured plasma levels of dd-cfDNA and correlated the levels with allograft rejection status ascertained by histology in 107 biopsy specimens. The dd-cfDNA level discriminated between biopsy specimens showing any rejection (T cell-mediated rejection or antibody-mediated rejection [ABMR]) and controls (no rejection histologically), P1% indicate a probability of active rejection

    Synchronized cycles of bacterial lysis for in vivo delivery

    Get PDF
    The pervasive view of bacteria as strictly pathogenic has given way to an ppreciation of the widespread prevalence of beneficial microbes within the human body. Given this milieu, it is perhaps inevitable that some bacteria would evolve to preferentially grow in environments that harbor disease and thus provide a natural platform for the development of engineered therapies. Such therapies could benefit from bacteria that are programmed to limit bacterial growth while continually producing and releasing cytotoxic agents in situ. Here, we engineer a clinically relevant bacterium to lyse synchronously at a threshold population density and to release genetically encoded cargo. Following quorum lysis, a small number of surviving bacteria reseed the growing population, thus leading to pulsatile delivery cycles. We use microfluidic devices to characterize the engineered lysis strain and we demonstrate its potential as a drug deliver platform via co-culture with human cancer cells in vitro. As a proof of principle, we track the bacterial population dynamics in ectopic syngeneic colorectal tumors in mice. The lysis strain exhibits pulsatile population dynamics in vivo, with mean bacterial luminescence that remained two orders of magnitude lower than an unmodified strain. Finally, guided by previous findings that certain bacteria can enhance the efficacy of standard therapies, we orally administer the lysis strain, alone or in combination with a clinical chemotherapeutic, to a syngeneic transplantation model of hepatic colorectal metastases. We find that the combination of both circuit-engineered bacteria and chemotherapy leads to a notable reduction of tumor activity along with a marked survival benefit over either therapy alone. Our approach establishes a methodology for leveraging the tools of synthetic biology to exploit the natural propensity for certain bacteria to colonize disease sites.National Institute of General Medical Sciences (U.S.) (GM069811)San Diego Center for Systems Biology (P50 GM085764)National Cancer Institute (U.S.). Swanson Biotechnology Center (Koch Institute Support Grant (P30-CA14051))National Institute of Environmental Health Sciences (Core Center Grant (P30- ES002109))National Institutes of Health (U.S.) (NIH Pathway to Independence Award NIH (K99 CA197649-01))Misrock Postdoctoral fellowshipNational Defense Science and Engineering Graduate (NDSEG) Fellowshi

    The C-terminal domain of p53 orchestrates the interplay between non-covalent and covalent poly(ADP-ribosyl)ation of p53 by PARP1

    Get PDF
    The post-translational modification poly(ADPribosyl)ation (PARylation) plays key roles in genome maintenance and transcription. Both non-covalent poly(ADP-ribose) binding and covalent PARylation control protein functions, however, it is unknown how the two modes of modification crosstalk mechanistically. Employing the tumor suppressor p53 as a model substrate, this study provides detailed insights into the interplay between noncovalent and covalent PARylation and unravels its functional significance in the regulation of p53. We reveal that the multifunctional Cterminal domain (CTD) of p53 acts as the central hub in the PARylation-dependent regulation of p53. Specifically, p53 bound to auto-PARylated PARP1 via highly specific non–covalent PAR-CTD interaction, which conveyed target specificity for its covalent PARylation by PARP1. Strikingly, fusing the p53-CTD to a protein that is normally not PARylated, renders this a target for covalent PARylation as well. Functional studies revealed that the p53–PAR interaction had substantial implications on molecular and cellular levels. Thus, PAR significantly influenced the complex p53–DNA binding properties and controlled p53 functions, with major implications on the p53-dependent interactome, transcription, and replication-associated recombination. Remarkably, this mechanism potentially also applies to other PARylation targets, since a bioinformatics analysis revealed that CTD-like regions are highly enriched in the PARylated proteome

    Flecainide Is Associated With a Lower Incidence of Arrhythmic Events in a Large Cohort of Patients With Catecholaminergic Polymorphic Ventricular Tachycardia

    Get PDF
    BACKGROUND: In severely affected patients with catecholaminergic polymorphic ventricular tachycardia, beta-blockers are often insufficiently protective. The purpose of this study was to evaluate whether flecainide is associated with a lower incidence of arrhythmic events (AEs) when added to beta-blockers in a large cohort of patients with catecholaminergic polymorphic ventricular tachycardia. METHODS: From 2 international registries, this multicenter case cross-over study included patients with a clinical or genetic diagnosis of catecholaminergic polymorphic ventricular tachycardia in whom flecainide was added to beta-blocker therapy. The study period was defined as the period in which background therapy (ie, beta-blocker type [beta1-selective or nonselective]), left cardiac sympathetic denervation, and implantable cardioverter defibrillator treatment status, remained unchanged within individual patients and was divided into pre-flecainide and on-flecainide periods. The primary end point was AEs, defined as sudden cardiac death, sudden cardiac arrest, appropriate implantable cardioverter defibrillator shock, and arrhythmic syncope. The association of flecainide with AE rates was assessed using a generalized linear mixed model assuming negative binomial distribution and random effects for patients. RESULTS: A total of 247 patients (123 [50%] females; median age at start of flecainide, 18 years [interquartile range, 14-29]; median flecainide dose, 2.2 mg/kg per day [interquartile range, 1.7-3.1]) were included. At baseline, all patients used a beta-blocker, 70 (28%) had an implantable cardioverter defibrillator, and 21 (9%) had a left cardiac sympathetic denervation. During a median pre-flecainide follow-up of 2.1 years (interquartile range, 0.4-7.2), 41 patients (17%) experienced 58 AEs (annual event rate, 5.6%). During a median on-flecainide follow-up of 2.9 years (interquartile range, 1.0-6.0), 23 patients (9%) experienced 38 AEs (annual event rate, 4.0%). There were significantly fewer AEs after initiation of flecainide (incidence rate ratio, 0.55 [95% CI, 0.38-0.83]; P=0.007). Among patients who were symptomatic before diagnosis or during the pre-flecainide period (n=167), flecainide was associated with significantly fewer AEs (incidence rate ratio, 0.49 [95% CI, 0.31-0.77]; P=0.002). Among patients with ≥1 AE on beta-blocker therapy (n=41), adding flecainide was also associated with significantly fewer AEs (incidence rate ratio, 0.25 [95% CI, 0.14-0.45]; P&lt;0.001). CONCLUSIONS: For patients with catecholaminergic polymorphic ventricular tachycardia, adding flecainide to beta-blocker therapy was associated with a lower incidence of AEs in the overall cohort, in symptomatic patients, and particularly in patients with breakthrough AEs while on beta-blocker therapy.</p

    Gut microbiota: Role in pathogen colonization, immune responses, and inflammatory disease

    Full text link
    Peer Reviewedhttps://deepblue.lib.umich.edu/bitstream/2027.42/138242/1/imr12567.pdfhttps://deepblue.lib.umich.edu/bitstream/2027.42/138242/2/imr12567_am.pd

    Effects of eight neuropsychiatric copy number variants on human brain structure

    Get PDF
    Many copy number variants (CNVs) confer risk for the same range of neurodevelopmental symptoms and psychiatric conditions including autism and schizophrenia. Yet, to date neuroimaging studies have typically been carried out one mutation at a time, showing that CNVs have large effects on brain anatomy. Here, we aimed to characterize and quantify the distinct brain morphometry effects and latent dimensions across 8 neuropsychiatric CNVs. We analyzed T1-weighted MRI data from clinically and non-clinically ascertained CNV carriers (deletion/duplication) at the 1q21.1 (n = 39/28), 16p11.2 (n = 87/78), 22q11.2 (n = 75/30), and 15q11.2 (n = 72/76) loci as well as 1296 non-carriers (controls). Case-control contrasts of all examined genomic loci demonstrated effects on brain anatomy, with deletions and duplications showing mirror effects at the global and regional levels. Although CNVs mainly showed distinct brain patterns, principal component analysis (PCA) loaded subsets of CNVs on two latent brain dimensions, which explained 32 and 29% of the variance of the 8 Cohen’s d maps. The cingulate gyrus, insula, supplementary motor cortex, and cerebellum were identified by PCA and multi-view pattern learning as top regions contributing to latent dimension shared across subsets of CNVs. The large proportion of distinct CNV effects on brain morphology may explain the small neuroimaging effect sizes reported in polygenic psychiatric conditions. Nevertheless, latent gene brain morphology dimensions will help subgroup the rapidly expanding landscape of neuropsychiatric variants and dissect the heterogeneity of idiopathic conditions

    Physical and functional interactions of the tumor suppressor protein p53 with poly(ADP-ribose) polymerase-1 and its enzymatic product poly(ADP-ribose)

    No full text
    Poly(ADP-ribosyl)ierung (PARylierung) ist eine wesentliche post-translationale Modifikation in Eukaryoten, die überwiegend von Poly(ADP-ribose) polymerase-1 (PARP1) ausgeführt wird. Sie spielt vor allem eine Rolle in der Signalkaskade von DNA Schäden, im Chromatin-Remodeling und in der Transkription. Proteine können kovalent PARyliert werden oder können nicht-kovalent mit poly(ADP-ribose) (PAR) interagieren. Das Tumorsuppressor protein p53 ist ein Substrat für kovalente PARylierung durch PARP1 und bindet auch nicht-kovalent PAR. Allerdings ist es unklar wie PARP1 p53 als Substrat für kovalente PARylierung erkennt und außerdem wie kovalente PARylierung und nicht-kovalente PAR Bindung in der Regulation von p53 zusammenwirken. In dieser Arbeit wurde p53 systematisch nach Regionen untersucht, die essenziell für nicht-kovalente PAR Bindung und für kovalente PARylierung von p53 sind. Eine bisher unbekannte PAR Bindestelle (Region 363-382) wurde in der hochbasischen und intrinsisch ungeordneten C-terminalen Domäne (CTD) von p53 entdeckt. Es stellte sich heraus, dass diese Stelle für die hauptsächliche PAR Bindung von p53 verantwortlich ist. Eine p53 Mutante konnte generiert werden, die kein PAR mehr bindet, indem vier entscheidende, basische Aminosäure zu Alanin ausgetauscht wurden. Interessanterweise ist dieselbe PAR Bindestelle auch essenziell für die kovalente PARylierung von p53 durch PARP1. Mehrere kovalente PARP1-vermittelte PARylierungsstellen konnten in der N-terminalen Transaktivierungsdomäne (TAD) von p53 identifiziert werden, wie E2, D7, E17 und E28. Wenn die TAD allerdings entfernt wird, kommt es immer noch zur kovalenten PARylierung. Nur wenn die nicht-kovalente PAR Bindung aufgehoben wird, wie durch das Entfernen der CTD, wird p53 auch defizient für kovalente PARylierung. Die p53 Mutante mit vier Aminosäuren-Substituierungen, die kein PAR mehr binden konnte, war ebenfalls defizient für kovalente PARylierung. Außerdem zeigte Auto-PARylierte PARP1 eine viel intensivere Interaktion zu p53, als unmodifizierte PARP1. Daher legen diese Ergebnisse nahe, dass nicht-kovalente PAR Bindung direkt die p53-PARP1 Interaktion vermitteln kann, sowie die Substraterkennung von p53 für kovalente PARylierung durch PARP1. Indem die CTD an ein Protein fusioniert wird, das normalerweise nicht PARyliert wird, wird dieses Protein ein Ziel von PARylierung. Dies bestätigt, dass die CTD der kritische Faktor für die Substraterkennung durch PARP1 ist. Bioinformatische Analysen zeigten, dass CTD-ähnliche Regionen im PARylierten Proteom stark angereichert sind. Dies legt nahe, dass ähnliche Mechanismen potentiell auch für die PARylierung von anderen Zielproteinen existieren. Zudem zeigten funktionelle Endpunktanalysen, dass nicht-kovalente p53-PAR Interaktion überwiegend die Sequenz-unabhängige, CTD-vermittelte DNA Bindung inhibiert. Andererseits wird die sequenz-spezifische DNA Bindung, vermittelt durch die zentrale DNA Bindungsdomäne (DBD) von p53, nur sehr gering durch PAR Bindung beeinflusst. Zelluläre Experimente haben gezeigt, dass PARylierung die transkriptionelle Aktivität von p53 beeinflusst, sowie die p53-Protein Interaktion und auch die p53-vermittelte Replikations-assoziierte Rekombination. Die Schlussfolgerung dieser Arbeit ist, dass nicht-kovalente PAR Bindung und kovalente PARylierung von p53 unzertrennlich miteinander verbunden sind und dass die CTD von p53 das Zentrum der Regulation von p53 durch PARylierung darstellt. Dieses Werk gibt eine mechanistische Erklärung wie p53 und potentiell andere Proteine zum Ziel von PARP1-vermittelter PARylierung werden.publishe

    Interactions of p53 with poly(ADP-ribose) and DNA induce distinct changes in protein structure as revealed by ATR-FTIR spectroscopy

    No full text
    Due to multiple domains and in part intrinsically disordered regions, structural analyses of p53 remain a challenging task, particularly in complex with DNA and other macromolecules. Here, we applied a novel attenuated total reflection Fourier transform infrared (ATR-FTIR) spectroscopic approach to investigate changes in secondary structure of full-length p53 induced by non-covalent interactions with DNA and poly(ADP-ribose) (PAR). To validate our approach, we confirmed a positive regulatory function of p53’s C-terminal domain (CTD) with regard to sequence-specific DNA binding and verified that the CTD mediates p53–PAR interaction. Further, we demonstrate that DNA and PAR interactions result in distinct structural changes of p53, indicating specific binding mechanisms via different domains. A time-dependent analysis of the interplay of DNA and PAR binding to p53 revealed that PAR represents p53’s preferred binding partner, which efficiently controls p53–DNA interaction. Moreover, we provide infrared spectroscopic data on PAR pointing to the absence of regular secondary structural elements. Finally, temperature-induced melting experiments via CD spectroscopy show that DNA binding stabilizes the structure of p53, while PAR binding can shift the irreversible formation of insoluble p53 aggregates to higher temperatures. In conclusion, this study provides detailed insights into the dynamic interplay of p53 binding to DNA and PAR at a formerly inaccessible molecular level.publishe

    Calcineurin stimulation by Cnb1p overproduction mitigates protein aggregation and α-synuclein toxicity in a yeast model of synucleinopathy

    Get PDF
    Abstract The calcium-responsive phosphatase, calcineurin, senses changes in Ca2+ concentrations in a calmodulin-dependent manner. Here we report that under non-stress conditions, inactivation of calcineurin signaling or deleting the calcineurin-dependent transcription factor CRZ1 triggered the formation of chaperone Hsp100p (Hsp104p)-associated protein aggregates in Saccharomyces cerevisiae. Furthermore, calcineurin inactivation aggravated α-Synuclein-related cytotoxicity. Conversely, elevated production of the calcineurin activator, Cnb1p, suppressed protein aggregation and cytotoxicity associated with the familial Parkinson’s disease-related mutant α-Synuclein A53T in a partly CRZ1-dependent manner. Activation of calcineurin boosted normal localization of both wild type and mutant α-synuclein to the plasma membrane, an intervention previously shown to mitigate α-synuclein toxicity in Parkinson’s disease models. The findings demonstrate that calcineurin signaling, and Ca2+ influx to the vacuole, limit protein quality control in non-stressed cells and may have implications for elucidating to which extent aberrant calcineurin signaling contributes to the progression of Parkinson’s disease(s) and other synucleinopathies. Video Abstrac
    • …
    corecore