text
stringlengths
1
32.7k
meta
dict
This is an application for a K08 Mentored Clinical Scientist Development Award from a clinically trained rheumatologist who now seeks to expand his career to become an independent physician scientist. Epstein- Barr virus (EBV) infection is more common in patients with systemic lupus erythematosus (SLE) than control subjects, suggesting that this virus plays an etiologic role in disease and/or that patients with lupus have impaired EBV-specific immune responses. The results of preliminary study of this proposal has demonstrated that patients with SLE have a defect in controlling latent EBV infection as evidenced by the increased EBV viral loads and altered EBV-specific T cell responses compared to healthy controls. Of interest, studies reported an increased frequency of herpes zoster due to reactivation of varicella zoster virus (VZV) in patients with SLE and increased EBV viral loads in peripheral blood of patients with rheumatoid arthritis (RA). These findings raise several questions: 1) whether the defect in controlling latent EBV infection is a unique finding in SLE or a global phenomenon in chronic immune-mediated diseases such as RA; and 2) whether this defect in SLE is limited to EBV or relavent to other latent viral infections such as VZV and cytomegalovirus (CMV). Next issues are the mechanisms for this defect and its clinical significance. To address these questions, three specific aims are planned. First, numerical and functional differences in EBV- and VZV-specific T cells between patients with SLE and healthy and diseased (RA) controls will be sought. Here, EBV- and VZV-specific CD4+ and CD8+ T cells in peripheral blood will be analyzed using MHC class I tetramers, flow cytometry following short-term in vitro stimulation and cytotoxic assay. EBV viral loads in peripherla blood will be measured using real-time PCR. Second, to find if the defect is secondary to immunosuppressive drugs and/or immune activation, EBV- and VZV- specific T cell responses and EBV viral loads in patients with new onset SLE will be determined using the same assays. Third, the effects of immunosuppressive therapies on EBV- and VZV-specific T cells and EBV viral loads in SLE will be assessed by serially measuring the frequency and function of EBV- and VZV-specific T cells and EBV viral loads before and after immunosuppressive therapies.
{ "pile_set_name": "NIH ExPorter" }
Translesion synthesis (TLS) DNA polymerases (Pols) help ensure the continued progression of the replication fork by promoting replication through DNA lesions. In the proposed studies, we will determine the roles of a number of human TLS Pols in promoting replication through a variety of DNA lesions induced by environmental pollutants and carcinogens and by cellular oxidative damage. In particular, we will test the hypothesis that replication through DNA lesions in human cells occurs via two distinct modes in which Pols, ?, ?, k, Rev1, and ? mediate predominantly error-free TLS and act in a highly specialized manner dependent upon the DNA lesion, whereas Pol? performs lesion bypass in a more generalized and error-prone manner. Further, we will test the hypothesis that the more generalized and error-prone role of Pol? emanates from its ability to insert a purine nucleotide (nt), preferentially an A, opposite DNA lesions via a protein-template-directed mechanism. To elucidate the genetic bases of error-free and mutagenic replication through DNA lesions in humans, we will carry out the following studies. In Aim 1, we will examine the contributions of various TLS Pols to error-free vs. mutagenic lesion bypass in human and mouse cells. The lesions to be studied include (6- 4) photoproduct induced by UV irradiation, 7,8-dihydro-8-oxogunaine (8-oxoG) generated from free-radical attack on guanine in DNA, 1,N6-ethenodeoxyadenosine (edA) generated from interaction of adenine with products of lipid peroxidation resulting from cellular oxidative damage and from exposure to chemical carcinogens, 1,N2-propano-2'-deoxyguanosine (PdG), a ring-closed form of acrolein generated from lipid peroxidation and which also is a ubiquitous environmental pollutant, and N2-dG adduct of the environmental carcinogen (+) anti-benzo[a]pyrene diol expoxide (BPDE). In Aim 2, biochemical studies will be done to examine the proficiency of Pol? in synthesizing DNA opposite (6-4) TT photoproduct, 8-oxoG, edA, PdG, and N2-dG BPDE. By steady-state kinetic analyses we will determine the catalytic efficiency of Pol? for inserting nucleotides opposite the DNA lesion and for carrying out the subsequent extension reaction, and biochemical studies will be done to test the hypothesis that Pol? inserts a purine nt opposite DNA lesions via a protein- template-directed mechanism. The genetic and biochemical studies we propose here are highly relevant for delineating the roles of TLS Pols in promoting error-free vs. mutagenic lesion bypass during replication, and for providing a comprehensive understanding of the genetic bases of mutagenesis and carcinogenesis induced by environmental and cellular DNA damaging agents in human cells. Our proposal for a predominantly error-free mode of TLS by Pols ?, ?, k, Rev1, and ? would predict a role for these Pols in cancer suppression, whereas a mutagenic mode of TLS by Pol? would predict a role for this Pol in enhancing genomic instability and carcinogenesis.
{ "pile_set_name": "NIH ExPorter" }
Many of the big, outstanding problems in biology involve complex, highly interactive processes. Nowhere is this more true than in the field of human health where disease severity can depend on social climate, individual behavior and previous exposures. Because of advances in biotechnology, it is now possible to investigate complex biological processes to a level of quantitative and functional detail unimaginable 20 or 30 years ago. However, the task is greatly complicated by the fact that the processes themselves cut across traditional scientific disciplines, leaving few specialized researchers fully prepared to answer them alone. There is clearly a great need for interdisciplinary research. What is less clear is how to achieve this at a level commensurate with the complexity of the problems we face. Central to our solution to this challenge is the Modeling Core. The Modeling Core is a research space, an arrangement of researchers around a nucleus of core fellows, and a culture of intellectual exchange all geared toward solving complex problems. The focus of the Modeling Core will initially be the projects of this center, each of which investigates complex interactions that underlie viral co- infection. With time, the core will expand to include new investigators tackling new biomedical problems, developing new methodological approaches to address them, and benefiting from the interdisciplinary dynamics within the core. We will achieve this vision through realizing the following aims: 1) establish the core: the people, its research space, the culture, and a strategy for assessment, 2) engage in integrative modeling that supports individual research projects and 3) promote outreach by extending core services and developing workshops. Interdisciplinary research addresses complex problems spanning multiple levels of biological organization, and it requires a deep exchange of ideas among fields to generate genuine insights. The Modeling Core is an innovative way to power interdisciplinary research by distilling the three critical features of this exchange: a shared language for connecting, a space for interaction, and a diversity of participants.
{ "pile_set_name": "NIH ExPorter" }
This project focused on the regulation of the N-methyl-D-aspartate (NMDA) peptide dynorphin. We demonstrated in the guinea pig hippocampal slice preparation that dynorphin can act as an agonist at two distinct receptors to regulate the function of NMDA receptors. The first site that dynorphin acts at in the hippocampal slice are the kappa2 opioid receptors. Dynorphin binding to this site results in an inhibition of the current that flows through the NMDA receptors. Prior to this discovery there was no known physiological function for the kappa2 opioid receptor. The second site that dynorphin acts at is the polyamine binding site located on the NMDA receptor. When dynorphin binds to this site the current flowing through the NMDA receptor is enhanced. The enhancement of the NMDA receptor-mediated current by dynorphin often results in neuronal damage and death in vitro. This finding is similar to the effect observed when dynorphin is injected onto the spinal cord of rats in vivo, suggesting that dynorphin's neurotoxic effects in vivo are mediated through the polyamine site of the NMDA receptor. It is well established that excessive activity in NMDA receptors may result in many different neuropathologies, including chronic pain. It is also well established that many of these pathologies are associated with elevated levels of endogenous dynorphin. Therefore, it is possible that endogenous dynorphin may play a role in enhancing the activity of NMDA receptors during pain. We are testing our hypothesis in a behavioral model of persistent pain. In rats whose right hind paw is inflamed by the injection of complete Freund's adjuvant, opioid agonists which act at the kappa2 opioid receptor inhibit the inflammation induced hyperalgesia when injected onto the spinal cord. The sensory profile of the non-inflamed paws are not altered by the kappa2 agonists. This finding is in contrast with the effects of other opioid agonists which alter the sensory profile of both the inflamed and the non-inflamed paws. The findings do, however, resemble results obtained by the injection of NMDA receptor antagonists, suggesting that the kappa2 receptors in vivo are regulating the NMDA receptors. Future work will focus on determining the mechanism by which kappa2 opioid receptors inhibit NMDA receptors and on the further characterization of the kappa2 opioid receptors and the polyamine binding site in the behavioral model.
{ "pile_set_name": "NIH ExPorter" }
This application will test the hypothesis that SP-A play a critical role in lung function during adenoviral mediated lung injury by modulated 1) host defense, 2) inflammatory responses, and 3) surfactant homeostasis, by interacting with surface receptors on respiratory epithelial cells and alveolar macrophages. We propose that SP-A serves complex regulatory roles in the lung, binding to cell surface receptors present on Type II cells and alveolar macrophages, influencing 1) binding, uptake and killing of microorganisms and 2) inflammatory responses of epithelial cells and alveolar macrophages to microorganisms. The application will utilize models in which the synthesis of SP-A is altered genetically, using SP-A/-, SP-A+/+ mice or by intratracheal instillation of purified mouse SP-A. SP-A will be replaced genetically, in distinct subsets of respiratory epithelial cells. SP-A deficient and replete mice will be exposed to adenovirus to determine the role of P-A in lung inflammation and function. The studies will discern the concentration dependent and region-specific role of SP-A in innate defense and in surfactant homeostasis in the lung. We will determine the molecular mechanisms controlling SP-A gene transcription in the normal lung and after adenoviral induced inflammation. The role of SP-A in the modulation of respiratory epithelial cells and alveolar macrophage responses to a pro- inflammatory stimulus induced by adenovirus will be assessed. We propose that SO-A interacts in complex ways with multiple receptors n target cells, including epithelial cells and alveolar macrophages. We will identify, clone and assess binding specificity of the SP-A/Clq receptor/binding protein that is proposed to mediate, at least in part, SPA-function. These studies will help clarify the role of SP-A in innate defense of the lung and provide the basis for future therapies to maintain endogenous or supply exogenous SP-A to prevent morbidity or bacterial infection.
{ "pile_set_name": "NIH ExPorter" }
The Data Coordinating Center (DCC) for the Dialysis Access Consortium (DAC) will coordinate the scientific and operational aspects of the two on-going DAC randomized placebo controlled clinical trials: the Fistula Trial and the Graft Trial, which will evaluate drug therapies that may reduce the failure and complication rate of arteriovenous grafts and fistulas in hemodialysis patients. The maintenance of adequate and safe access to the circulation is critical to the hemodialysis treatment of patients with ESRD. The specific aim for the Fistula Trial is to determine if the 6-week administration of the anti-platelet drug, clopidogrel, increases the patency rate of newly placed fistulas. A secondary aim is to determine if clopidogrel increases the number of fistulas that are suitable for dialysis. The specific aim for the Graft Trial is to determine if continuous administration of Aggrenox (containing dipyridamole and a small amount of aspirin) prolongs the primary unassisted patency in newly constructed grafts. Systems for data acquisition (via a secure web-based data entry), data management, and quality control are being used. The DCC provides training to clinical center staff, and arranges and actively participates in meetings and conference calls of the Steering Committee. During the recruitment and follow-up phase of each trial, the DCC monitors patient recruitment and compliance and uses the database management system to assure accurate and complete collection of trial data. The DCC also coordinates the supply of study drugs and collection of stored biospecimen samples. An inquiry system is used to resolve data discrepancies. Trial progress is presented in reports to the clinical centers, Steering Committee, and the Data and Safety Monitoring Board. Statistical and interim analyses are performed for each trial with final analyses completed at the end. Trial results will be reported and published. At the end of the study, the data will be archived at the NIDDK Data Repository. If either or both of the study drugs are found to be beneficial, the morbidity, and possibly mortality, of hemodialysis patients will improve substantially. The health care cost of maintaining adequate vascular access should also decrease from its current rate of over one billion dollars per year.
{ "pile_set_name": "NIH ExPorter" }
The goal of this project is to develop a commercially and scientifically successful software system, named Teli Us Medicine, designed to meet current and future needs in palliative and hospice care markets, addressing the management of pain and other symptoms to improve quality of life. A core purpose for Tell Us Medicine is data-based communication with, and between, patients and their families, professional care providers, administrators, and researchers. Based on HL7, CDISC, and caBIG models and standards, and an existing health science-architected web technology platform , the data infrastructure for Tell Us Medicine will enable integration, interoperability, and ongoing adaptability to evolving scientific knowledge, best practices, and innovation. The system is purely web-based, so that the software can be accessed and used with any web-enabled devices, including devices such as smartphones and tablets. The focus in Phase II is standard personal computers in home and office settings.
{ "pile_set_name": "NIH ExPorter" }
The objective of this research is to analyze normal and abnormal mechanisms of development through use of mutant genes of the mouse. The mutations to be studied include: (1) tight skin (Tsk), a dominant mutation causing extensive hyperplasia of the loose connective tissue, enlarged heart and lungs, and abnormally long cartilages and long bones; (2) motheaten (me) a recessive mutation causing extensive skin lesions, severe pneumonitis, and early death, and showing evidence of an immune deficiency. The methods will include light and electron microscopy and the experimental methods appropriate for immunological studies.
{ "pile_set_name": "NIH ExPorter" }
PROJECT SUMMARY As the primary cellular mediators of hemostasis, platelets are optimized to limit bleeding through rapid adhesion, secretion and aggregation responses at sites of endothelial injury. Platelets also adhere to dysfunctional endothelium, where they secrete proinflammatory molecules and form aggregates with leukocytes to progress vascular inflammation in a manner relevant to the pathogenesis of chronic diseases, including atherosclerosis. Ongoing efforts aiming to understand and target platelet activities specific to disease have characterized a spectrum of platelet functional phenotypes associated with inflammatory, thrombotic and other conditions. Despite the identification of key molecular alterations that highlight differences between these phenotypes, it remains unclear how different platelet phenotypes develop, how they should be defined, and, ultimately, how they should be targeted. We hypothesize that platelet hemostatic, inflammatory and other phenotypes are determined by the systematic activation of intracellular signaling pathways and effectors that result in specific platelet functional outputs in response to physiological context. We aim to systematically define intracellular signaling events that progress platelet adhesion (Aim 1), secretion (Aim 2) and aggregation (Aim 3) in hemostatic programs and to determine how these responses mechanistically differ in the context of vascular inflammation. We will engage these studies through the use of a high-throughput, proteomics-based workflow that measures and maps intracellular signaling events and pathways underlying platelet function in specific experimental and physiological contexts. We now use this set of proteomics, computational and cell biological tools to build pathway maps intracellular signaling relations in platelet activation programs. In this proposal, we use this first-in-class pathway mapping methodology together with other physiological and systems biology tools to address how platelet signaling programs specify platelet phenotypes favoring hemostatic and inflammatory responses. Ultimately, this work will generate knowledge as well as a conceptual means to define and understand systems level mechanisms of platelet regulation in hemostasis as well as in inflammation and the manifestation of disease.
{ "pile_set_name": "NIH ExPorter" }
Studies are being performed to assess the role of nonspecific immune responses in the acquisition of age-dependent resistance to Sindbis virus encephalitis of mice. In vivo studies, on the reticuloendothelial cell system, natural killer cells, interferon, as well as the complement system are being evaluated in young, susceptible mice, and older resistant mice. In vitro studies are focused on the activation of the complement system (classical and alternative pathways) by Sinbis virus and the biological consequence of such activation.
{ "pile_set_name": "NIH ExPorter" }
Ammonia is an irritant gas and even at sub-ppm concentrations has been found to be associated with respiratory disease. Occupational exposures to ammonia are encountered in commercial refrigeration, janitorial work and animal agriculture. Occupational exposure to ammonia have been linked to adverse health outcomes at levels well below the current PEL of 50 ppm, however understanding the specific role that chronic exposures to low levels of ammonia play in these adverse health outcomes is hampered by an almost complete absence of data on personal occupational exposures to ammonia at low-ppm and sub-ppm concentrations. This data gap exists in large part because current ammonia monitoring equipment is expensive, is often not portable, and consequently is poorly suited to personal and community exposure monitoring. The objective of this proposed project is to develop a low cost ammonia sensor array that can be integrated with sensors for particle mass and other gases, for use in exposure assessments of ammonia concentrations in workplaces such as animal feeding operations. The specific aims of the project are: 1) Fabricate sub- millimeter scale ammonia sensors with sub-ppm sensitivity based on reduced graphene oxide chemiresistors; 2) Optimize the reduced graphene oxide sensor array for selectivity and sensitivity; 3) Incorporate the new ammonia sensor element into an existing portable monitoring device; and 4) Evaluate performance of the portable monitoring device in the field. Our proposed research addresses several aspects of NIOSH' National Occupational Research Agenda (NORA). The novel ammonia sensor, incorporated into a portable exposure monitoring device addresses health risks in the Agriculture, Forestry and Fishing Sector, and the Services Sector, and cross sector programs in Exposure Assessment, Respiratory Diseases and Total Worker Health. The tools developed in this proposal are not specific to ammonia sensors but are broadly intertwined with efforts in advancing the technology to use low cost sensors for exposure measurements. The outputs from this project are 1) assessments of novel ammonia sensor technologies for use in occupational hygiene 2) development of a new ammonia monitor with sub-ppm sensitivity for continuous ammonia monitoring 3) novel designs for adapting the low cost sensors for use in a personal exposure monitor 4) improved informatics tools for processing and analyzing data applicable to a low cost monitoring network.
{ "pile_set_name": "NIH ExPorter" }
There is a scientific gap in our current understanding of how formulation and raw material variables used to manufacture poly(lactic-co-glycolic acid) (PLGA) microspheres encapsulating peptides lead to differing levels of peptide-polymer interactions and peptide acylation during encapsulation, storage, and release in vitro and in vivo. Batch-to-batch variation and unpredictable acylation levels could lead to differences in pharmacokinetics (PK) and loss of bioequivalence of generic long-acting release (LAR) formulations seeking to mimic LAR products. Such differences in bioequivalence may lead to differences in safety and efficacy. A key factor leading to the loss of parent drug is the poorly defined interaction between the peptide and the polymer, which has been shown to be a precursor to the acylation reaction. For example, salt formation between the cationic and acylation-labile octreotide, found in the Sandostatin LAR (SLAR) microsphere product, with the ionized carboxylic acid end-groups of PLGA has been shown to be necessary for acylation to occur. We hypothesize the key underlying time-dependent factors responsible for differences in peptide-polymer interactions and acylation for microspheres of similar compositions include: (a) microclimate pH in the microspheres affecting ionization of polymer carboxylic acids necessary to bind to a cationic peptide; (b) the extent to which the polymer allows penetration of the peptide into the polymer phase, and (c) the architecture of the polymer (star vs. linear) and end-capping that affect the strength and type of intermolecular forces responsible for peptide- polymer interactions. The following tasks will be developed to test our hypothesis and close the aforementioned scientific gap: 1. Develop techniques for assessing peptide-polymer interactions and peptide acylation impurities with the reference product, SLAR; 2. Reverse engineer the composition of SLAR; 3. Create Q1/Q2 formulations for the SLAR as a function of microencapsulation conditions, and determine product attributes relative to SLAR. 4. Create microsphere formulations with virtually the same composition of SLAR but with different PLGA architecture and molecular weight, and determine product attributes; 5. Compare peptide-polymer interactions, acylation, erosion behavior and release kinetics in all formulations to identify key formulation and polymer architecture controlling differences in peptide acylation and release kinetics in vitro; and 6. Verify in vitro differences observed in Task 1 and 5 by assessing key formulations for PK after intramuscular injection and direct measurements after recovery from a novel in vivo cage model. We will apply a wide range of novel experimental approaches developed by our group over the last 20 years to interrogate the peptide-polymer interactions necessary to initiate octreotide acylation in PLGAs. We will further combine this approach with a large number of rigorous and new in vitro assays to test formulations with relevance to future regulatory guidance to generics and verify their relevance to the not yet predictable in vivo environment.
{ "pile_set_name": "NIH ExPorter" }
End-stage renal disease (ESRD) was inevitably fatal until the development of an external shunt together with the use of dialysis technology. According to the NIH, the incidence of treated ESRD in the US is rising 7.8% per year. Over 200,000 ESRD patients are presently being treated with hemodialysis through a graft/vein shunt. Current experience has shown that these grafts typically fall in 14-19 months. Clinically, most graft failures are secondary to thrombosis resulting from stenosis at the graft/vein anastomosis. Stenosis is the result of a cellular proliferation similar to that observed following other vascular interventions, including angioplasty and synthetic bypass graft placement. Similar intimal hyperplasia responses to vascular interventions (e.g. angioplasty) have been recently treated with ionizing radiation. Nearly all of the methods thus far proposed treat arterial vessels from the inside. Stenotic lesions are frequently eccentric within the vessel lumen, and proliferation is believed to be initiated in the smooth muscle cells near the adventitial layer of the vessel. It is proposed that an optimum prophylactic treatment for the threatened stenosis would deliver therapeutic levels of ionizing radiation over an extended period of time via a device that is positioned around the vessel/graft junction at the time of graft implant surgery. An external "wrap-around" source of radiation provides far greater spatial uniformity of dose in the treatment zone than an internal source. The selection of backing is important for minimizing dose to other tissue and to the physician. Only a single procedure is required to install both the graft and the wrap. PROPOSED COMMERCIAL APPLICATIONS: End-stage renal disease is treated with dialysis technology using an external shunt for access. The shunt graft frequently fails within a year due to low blood flow caused by stenosis. This proposal describes a method for inhibiting stenosis, significantly reducing revision surgeries and trauma to the patient.
{ "pile_set_name": "NIH ExPorter" }
A general modelling program (MKMODEL) has been developed for the PROPHET computer system which allows combined pharmacokinetic and pharmacodynamic simulation and curve fitting. The effect of heart rate on the QT interval in dogs has been shown to be a hyperbolic function. The effect of quinidine is additive and described by a linear model.
{ "pile_set_name": "NIH ExPorter" }
The Department of Radiology at UCSD has implemented a Small Animal Imaging Resource (SAIR) to support cancer research at UCSD and the San Diego bioresearch community. The SAIR is located in the vivarium at the Rebecca and John Moores UCSD Comprehensive Cancer Center (CC) on the La Jolla campus. This facility supports the efforts of the ICMIC investigators and the UCSD cancer research investigators and then Campus and city-wide efforts. Some non-UCSD potential users include: Sidney Kimmel CC, Scripps, Burnham and Salk Institutes, and the San Diego biotech companies. The UCSD SAIR will provide MRI, optical, CT, ultrasound, High-Resolution planar gamma imaging, and PET of rodents, as well as high-resolution digital autoradiography and fluorescent imaging of thin whole body rodent sections. Except for MRI, all modalities are at one site adjacent to the CC vivarium. Support and expertise includes optical imaging, animal support, image computation, MR & optical hardware and software, diagnostic agent chemistry, radiochemistry including a cyclotron, analytical chemistry, kinetic modeling, informatics, histology, and a vascularized solid tumor model to cost-effectively screen imaging paradigms. Our primary emphasis will be the acquisition of images for kinetic modeling. For this reason the kinetic modeling service will develop automated data reduction schemes. Our previous experience with the use of kinetic modeling for receptor density measurement from clinical studies has convinced us that the data reduction path from the image data set must be completely automated. There are two reasons for this: first is high throughput, and second and more important is the absence of operator intervention and the elimination of operator bias. This latter quality is essential for rigorous statistical analysis (by the ICMIC Biostatistics Service) of the imaging and biochemical/physiologic metrics. High-resolution ultrasound imaging will permit quantification of tumor size as well as measurement of left ventricular volume for recovery correction of PET sampled input functions. Most importantly, it will permit the use of in situ tumor models. A key contribution of ultrasound to cancer research in mice is the ability to image in sterile fashion and to do so without the need for anesthesia. This should permit frequent monitoring of tumor growth or regression following interventions. Members of the ICMIC Imaging Core will interact closely with colleagues of the Animal Models & Care and the IMCIC Biostatistics Service. Our goal will be the identification of the most effective imaging model and the efficient use of animals. This will be accomplished by weekly meetings of at least one member from each Core. These meetings will include: 1) meetings with new Pis to develop new imaging projects prior to composing new IACUC protocols, and 2) the review of completed IACUC protocols prior to IACUC submission.
{ "pile_set_name": "NIH ExPorter" }
The intracellular level of cholesterol and cholesteryl esters controls the level of the microsomal enzyme 3-hydroxy-3-methylglutaryl Coenzyme A (HMG-CoA) reductase and thereby regulates the rate of cholesterol synthesis. The long-term goal of these investigations is to elucidate the molecular and biochemical mechanisms through which cholesterol controls the level of HMG-CoA reductase. Our approach is to establish the site at which exogenous cholesterol suppresses HMG-CoA reductase synthesis in established cell cultures of animal and human origin. Normal liver cell cultures adapted for growth in chemically defined medium and regulatory variants defective in regulation by oxygenated sterols or cholesterol are employed in these investigations. The site of cholesterol action will be established through immunochemical studies of HMG-CoA reductase synthesis. The effects of cholesterol on the level of HMG-CoA reductase specific messenger RNA, on the efficiency of translation of HMG-CoA reductase mRNA in vivo and on the integration of newly synthesized HMG-CoA reductase into the endoplasmic reticulum will be assessed. The HMG-CoA reductase mRNA content of RNA preparations will be determined by indirect immunoprecipitation of the reductase synthesized in a cell free protein synthesizing system derived from nuclease treated rabbit reticulocyte lysate. The location, number and size of HMG-CoA reductase synthesizing polyribosomes will be determined in iodinated antibody binding studies and by mRNA assay and the possible occurence of translation control and a hydrophobic "signal peptide" on newly synthesized reductase will be examined. The regulatory variants produced can serve as models for clinical syndromes in which defective cholesterol regulation of HMG-CoA reductase results in serum hypercholesterolemia and atheroschlerosis. Cholesterol regulation of HMG-CoA reductase synthesis represents a favorable system for investigation of the mechanisms governing gene expression and protein synthesis in animal and human cells.
{ "pile_set_name": "NIH ExPorter" }
The primary focus of the section is to further our understanding of the molecular basis of signaling between G protein coupled receptors and voltage gated ion channels in neurons using electrophysiological, molecular, and imaging techniques. A current project involves the investigation of a recently de-orphanized G-protein coupled receptor termed GPR41 or FFA3 in rodent sympathetic neurons. These receptors use short-chained fatty acids (SCFA; e.g., acetate and propionate) as endogenous ligands but little else is known. We have discovered that GPR41 is natively expressed and functionally coupled to calcium channels in sympathetic neurons (primarily the celiac/superior mesenteric ganglia). We have also generated evidence that receptor expression is highest in paravertebral and prevertebral sympathetic neurons. Finally, our data indicate that a ketone body, beta-hydroxybutyrate, acts as an agonist at this receptor. These findings are potentially important for several reasons. First, GPR41 may be expressed in neurons that innervate adipose tissue and thus be an important target for anti-obesity drugs. Second, we proposed that beta-hydroxybutyrate may serve as the primary endogenous ligand for sympathetic ganglia. This finding has implications for conditions in which plasma beta-hydroxybutyrate is elevated such as diabetic ketoacidosis or ketogenic diets. Third, acetate is a major metabolite of ethanol. Hence, GPR41 may be involved in both the response to ethanol (especially hangover symptoms) and addiction. It is possible that polymorphisms in GPR41 and primate-specific segmental duplication GPR42 contribute to susceptibility to alcoholism in humans. Fourth, as GPR41 is likely capable of detecting ethanol indirectly via blood acetate levels, it may be possible to leverage this property to interrupt neural pathways contributing to addiction in model organisms using genetic techniques. Won Y-J, Lu VB, Puhl HL, Ikeda SR (2013) Beta-Hydroxybutyrate Modulates N-Type Calcium Channels in Rat Sympathetic Neurons by Acting as an Agonist for the G-Protein-Coupled Receptor FFA3. J Neurosci 33:1931419325. As a logical extension to this study, we are examining the sequence of the human FFA3 gene. FFA3 is segmentally duplicated in primates including humans giving rise to a second gene termed GPR42. Our preliminary work indicates extreme variability in the open reading frames of both FFA3 and GPR42. Moreover, we find that GPR42 is both transcribed and functional when heterologously expressed thus challenging long held beliefs that GPR42 is non-functional or a pseudogene. Finally, we have generated evidence for a common structural variation in this region that results in a GPR42 copy number variation. Together, these results indicate unexpected variability in FFA3/GPR42 genotype and copy number that could potential impact functions dependent on this receptor class. A second project investigated the RGK proteins (Gem, Rad, Rem1, and Rem2), members of the Ras superfamily of small GTP-binding proteins that interact with calcium channel beta subunits to modify voltage-gated calcium channel function. In addition, RGK proteins affect several cellular processes such as cytoskeletal rearrangement, neuronal dendritic complexity, and synapse formation. To probe the phylogenetic origins of RGK proteincalcium channel interactions, we identified potential RGK-like protein homologs in genomes of genetically diverse organisms from both the deuterostome and protostome animal superphyla. RGK-like protein homologs cloned from Danio rerio (zebrafish) and Drosophila melanogaster (fruit flies) expressed in mammalian sympathetic neurons decreased calcium current density as reported for expression of mammalian RGK proteins. Sequence alignments from evolutionarily diverse organisms spanning the protostome/ deuterostome divide revealed conservation of residues within the RGK G-domain involved in RGK proteincalcium channel beta subunit interaction. In addition, the C-terminal eleven residues were highly conserved and constituted a signature sequence unique to RGK proteins but of unknown function. Taken together, these data suggest that RGK proteins, and the ability to modify calcium channel function, arose from an ancestor predating the protostomes split from deuterostomes approximately 550 million years ago. Puhl HL, Lu VB, Won Y-J, Sasson Y, Hirsch JA, Ono F, Ikeda SR (2014) Ancient origins of RGK protein function: modulation of voltage-gated calcium channels preceded the protostome and deuterostome split. PLoS ONE 9:e100694.
{ "pile_set_name": "NIH ExPorter" }
Oxidant injury by hyperoxia of rat lung initially results in alveolar epithelium and capillary endothelium injury with influx of inflammatory cells and subsequent progression to fibrosis. Although the critical roles of soluble factors produced by inflammatory cells in the recruitment of fibroblasts in initiating fibrogenesis are at present the focus of extensive investigation, few studies have been directed towards examining the earlier molecular events that occur prior to any observable alterations in lung pathology or physiology. There is even less known on the effects of aging on the ability of cells to respond to oxidative stress. We hypothesize that the genes expressed acutely by lung cells in response to oxidative stress depend on the extent of cellular damage and predict the subsequent progression to lung fibrosis. One or another of these early genetic responses may become defective with age and be associated with the irreversible decline in lung function. Here we propose to study the variety of genetic responses (anti-oxidants, heat shock proteins and DNA damage) to increasing levels of oxidative damage in the whole lung to determine the specificity of the response and whether these responses are age-related. We will localize these genetic responses at the histological level by in situ hybridization since the cellular response in the lung is likely to involve specific sites and cell types. Finally, attempts will be made to relate the production of various mediators involved in lung fibrosis with one of the acute or early cellular responses to stress. We expect that those studies will show that a certain level of damage must be achieved before the mediators of irreversible lung damage will be induced. A comparison of the responses in various aged animals will establish whether protective genetic responses are reduced with aging.
{ "pile_set_name": "NIH ExPorter" }
The endocannabinoid system consists of two cannabinoid receptors CB1 and CB2, which are G-protein coupled receptors and activated by mainly lipidic compounds. CB1 is a desirable therapeutic target due to its involvement in pathways related to addictive disorders and obesity. Hemopressin (HP), a nine-amino acid- peptide derived from the a chain of hemoglobin, has been shown to have selective inverse agonist activity against the CB1 receptor. In spite of hemopressin's tremendous potential as a safe and effective therapeutic, its further development has been hampered due to the variability of synthetic hemopressin in pharmacological assays. We hypothesize that the variability of hemopressin's effects in biological assays is due to the tendency of this peptide to form self-assembled nanostructures in solution under physiologically relevant conditions. Accordingly, we propose two specific aims: 1) To design and synthesize synthetic conjugates and analogs of hemopressin and conduct detailed nuclear magnetic resonance spectroscopy and transmission electron microscopy experiments to assess their aggregation and self-assembly properties. 2) To assess the affinity and efficacy of the newly designed conjugates and analogs of HP towards CB1 using four different assay systems: i) CB1 receptor binding assay, ii) [35S]GTPgS binding assay to monitor G-protein activation, iii) receptor-activated inhibition of cellular cAMP, and iv) b-arrestin recruitment. The central objective of this application is to evaluate the impact of hemopressin's ability to form nanofibrils on its pharmacological properties. Self-assembling biological peptides such as b-amyloid have profoundly improved our understanding of many aspects of neurobiology. Similarly, if we are able to show that self-assembly and aggregation of HP modulate its pharmacological activity, the finding will have promising therapeutic applicability in drug abuse research. PUBLIC HEALTH RELEVANCE: The endocannabinoid system refers to a group of neuromodulatory lipids and their receptors that are involved in a variety of biological processes, including addiction, appetite, memory, pain sensation, cognition and cancer cell proliferation. The endocannabinoid system consists of two cannabinoid receptors CB1 and CB2. Hemopressin is a hemoglobin-derived peptide that has been shown to act as a selective inverse agonist for the CB1 receptor, which is predominantly expressed in the motivational circuitry of the brain. CB1 is a desirable therapeutic target due to its involvement in pathways related to addictive disorders and obesity. Hemopressin is a potential drug candidate but its pharmacological activity has not been consistent and reproducible, possibly due to its propensity to form aggregates under physiological conditions. The goal of this study is to investigate the aggregation properties of hemopressin and develop analogs and/or conjugates of this peptide as therapeutic leads that show reproducible pharmacological profiles.
{ "pile_set_name": "NIH ExPorter" }
Recent studies from our laboratory, utilizing cell-specific gene targeting, indicate that collecting duct (CD)-derived endothelin-1 (ET-1) is an important regulator of systemic blood pressure and renal Na and water excretion. Based on this work, we hypothesize the following: CD ET-1 production is increased in conditions necessitating enhanced Na and water excretion and reduced blood pressure. Activation of CD ETB receptors and medullary interstitial cell ET receptors increases medullary nitric oxide and PGE2 production, while activation of CD ETA receptors enhances superoxide formation which reduces nitric oxide. Nitric oxide and PGE2 inhibit CD Na and/or water reabsorption and dilate medullary vasa recta. The resultant Na and water excretion limits hypertension in the setting of Na loading or mineralocorticoid excess. CD ET-1 may play a different role in angiotensin II hypertension due to angiotensin II down-regulation of the medullary ET-1 system. This system is also crucial in mediating vasopressin escape. Overall, the CD ET-1 system is essential in controlling blood pressure under normal physiologic as well as pathologic conditions. Mice with CD-specific knockout of ET-1, ETA receptor, ETB receptor, or both ETA and ETB receptors will be used. The mechanisms of ET-1 regulation of renal function and blood pressure under normal physiologic circumstances will be studied. This includes determination of whether CD-derived ET-1 acts in an autocrine and/or paracrine fashion, whether ETA and ETB receptors have opposing effects on Na and water excretion and blood pressure, whether and how CD-derived ET-1 regulates medullary blood flow, significance and mechanisms of CD-derived ET-1 interaction with nitric oxide, PGE2 and superoxide systems, and whether CD-derived ET-1 causes long-term changes in CD Na and water transporter expression. In addition, CD-derived ET-1 regulation of blood pressure and renal function under salt and/or water retaining conditions will be examined. This will include determination of the role of CD-derived ET-1 in controlling blood pressure and/or maintaining renal function in DOCA/salt hypertension, angiotensin II hypertension, and AVP excess. These studies will provide information on the role of CD-derived ET-1 in controlled blood pressure and renal function under normal physiologic as well as pathologic conditions. Such information is important in understanding intrarenal mechanisms responsible for hypertension and salt and water retention.
{ "pile_set_name": "NIH ExPorter" }
Aberrant Notch signals have been linked to cancer and other human diseases, which has motivated the pharmaceutical industry to develop agents inhibiting Notch signaling. However, studies in the mouse predicted that these agents would be toxic to various organ systems. Confirmed in humans are toxicities to the gastrointestinal track and skin, the latter leading to elevated rates of non-melanoma skin cancer. We have shown that targeting Notch1 specifically will result in vascular tumors in addition to increasing cancer rates in the skin; if this will occur in humans it will limit the therapeutic potential of anti-Notch1 therapies in a chronic disease setting. Recently, we and our collaborators reported that dimerization of active Notch molecules is an important step in the activation of some target genes and is required for the oncogenic activity of Notch1 in T-cell leukemia (T-ALL). The absolute dependence of T- ALL on Notch dimerization provides a novel mechanism-based therapeutic avenue. However, the biological consequences of targeting this aspect in Notch signaling are currently unknown due to novelty of the observation and the lack of tools that can differentiate dimerization-dependent from -independent targets. We were fortunate to be awarded ARRA funds to develop a new technology that can interrogate target selection by different multi-member complexes using complementing fragments of the E. coli DNA Adenine methyltransferase (DAM). It is an ideal novel method to identify Notch dimerization dependent targets. In this application we propose to facilitate drug development efforts for 'dimer-busting' therapeutics by defining the therapeutic window (the spectrum of untoward effects in vivo), by identifying dimerization dependent targets (should this window prove to be too narrow or the drug discovery efforts too difficult) and by utilizing our dimer-sensitive assay and high throughput screening to identify drug leads.
{ "pile_set_name": "NIH ExPorter" }
Producing potent high-affinity antibodies that can neutralize target pathogens remains a central goal for vaccine research. While a great deal is known about antibody molecules themselves, still too little is understood about the programming of high-affinity memory B cells that produce them. Many promising vaccine antigens fail to elicit protective antibodies using contemporary vaccine formulations. Thus, major gaps remain in our basic understanding of antibody affinity maturation and how to enhance poor immunogenicity using rational protein vaccination. Research Focus: Adjuvants are required to induce potent antibody responses using protein-based vaccine formulations. Follicular helper T (TFH) cells are a new class of immune regulator specialized to control multiple stages of memory B cell development. Here, we examine the innate cellular and molecular regulators of antigen-specific TFH development at the point of initial priming and following the vaccine boost. Specific Aims: Using new single cell strategies, we will dissect the molecular components of regulatory programs within peptide MHCII-expressing dendritic cell (DC) that control antigen-specific TFH programming in vivo (SA-1). Upon antigen recall, we now reveal that memory B cells form secondary GC reactions, re-diversify their expressed BCR, with evidence for ongoing clonal selection. Importantly, addition of secondary adjuvant at the vaccine boost enhanced affinity maturation within the memory B cell response. These recent findings alter the prevailing view of BCR affinity maturation and indicate new innate mechanisms for regulating high-affinity B cell memory (SA-2). Impact: We use polyclonal murine models of protein vaccination with state-of-the-art single cell analyses of antigen-specific immune function. These high-resolution studies of model antigens have the capacity to shift the basic conceptual framework that surrounds existing vaccine paradigms. Importantly, these basic new principles and assays can be used to re-design contemporary vaccine formulations that optimize high-affinity B cell immunity to more complex antigens.
{ "pile_set_name": "NIH ExPorter" }
The primary goal of this project is to build and test a prototype instrument for high throughput protein crystallography; it would automate the monitoring and storage of crystallization experiments form their initial "set up" (typically on a robot) through he final extraction for structure determination. We call this instrument a "Protein Crystallization Monitor Station" (or PCMS). It would store and monitor large number of crystallization experiment (in exchangeable cassettes of 180 microplates containint 17,280 individual experiments) at constant temperature and humidity. Crystallizations would be monitored by digital image capture; the digital image would be initially screened by machine vision software that would significantly reduce the number of images requiring human interpretation. The software associated with the PCMS would be integrated with the XtalGrow software developed in the Principal Investigator's laboratory. PROPOSED COMMERCIAL APPLICATIONS: This project will develop an automated instrument that would facilitate protein crystallization for both high-throughput and conventional protein crystallization efforts.
{ "pile_set_name": "NIH ExPorter" }
A well-established finding in cognitive aging is that older adults do not perform as well as young adults on tests of episodic memory. Episodic memories are contextually-specific, such as events that occurred at a particular place and time. Indeed, memory deficits have been found to be nearly twice as large for contextual details than for content items. The exact mechanism underlying this memory deficit remains unclear. One explanation for older adults'relatively poor performance is an impairment in the process of both creating and retrieving links, or relationships, among the separate contextual features of a to-be remembered episode. An alternative viewpoint posits that the impairment evident in episodic memory is part of a more general age-related decline in strategic, controlled processes. Such processes include the intentional manipulation, organization, or evaluation of features or contextual attributes and, especially, the conscious retrieval of contextual or relational information. According to this view, automatic or unintentional processes remain relatively unimpaired in aging. A way to differentiate between these alternative viewpoints is to examine implicit relational memory. Implicit memory refers to non-conscious, unintentional influences of memory - when some aspect of a previous experience influences or facilitates behavior in a new, seemingly unrelated situation. If a deficit in binding mechanisms at encoding can explain later episodic memory impairment, then such a deficit should be evident using both tests of both implicit and explicit retrieval. Alternatively, if older adults are impaired in strategic, controlled processing of relational information, then an age-related deficit should not occur with implicit testing. The current project proposes three studies aimed at determining: 1) whether the typical finding of age-related impairment in relational processing can be generalized to implicit memory, using two complementary paradigms, and 2) whether older and younger adults exhibit similar medial temporal lobe (MTL) activations during encoding and implicit relational retrieval, using functional magnetic resonance imaging (fMRI). Critically, the proposed experiments are aimed at resolving two competing viewpoints;thus, results in either direction are intended to be informative for the greater theoretical context. The focus of this project is aligned with the broader goal of conducting clinically applicable research related to the cognitive neuroscience of aging. Understanding the nature of this deficit in relational processing is not only of theoretical importance, but also has critical implications for older adults'day-to-day memory function. The ability to bind elements together into a rich, contextual representation is what allows for successful and coherent memories of everyday events.
{ "pile_set_name": "NIH ExPorter" }
Project Summary Scholarly and public interest in fatherhood has increased in recent decades, and this interest has coincided with revised cultural perceptions about what makes a good father. In addition to serving as breadwinners and protectors, fathers are also expected to be engaged in their children's lives. These competing demands contribute to work-family conflict, yet scholars have largely ignored one important work-family policy: paternity leave. Paternity leave can provide fathers with opportunities to learn parenting skills, bond with their child, and strengthen father identities while also maintaining employment. Thus, paternity leave can enable fathers to adhere to the expectations of both traditional and new fatherhood by encouraging fathers to contribute resources that facilitate child development. Paternity leave-taking may also alleviate mother's stress and work- family conflict by offering families a respite that may lead to increases in co-parents' relationship quality and mother's well-being. As a result, increased access to paternity leave may help to reduce gender inequality by allowing mothers to (re)enter the labor force sooner if they so desire, improving mothers' earnings, and reducing mothers' share of domestic tasks. Unfortunately, little is known about why fathers do (or do not) take paternity leave and what influence paternity leave-taking may have on families. Using longitudinal data from three national datasets (the National Longitudinal Study of Youth 1997, the Early Childhood Longitudinal Study-Birth Cohort, and the Fragile Families and Child Wellbeing Study), the proposed study examines the relationship between paternity leave and family well-being by focusing on who takes paternity leave, and whether and how paternity leave is associated with indicators of short- and long-term family well-being. We address three specific aims in this study: (1) specify patterns of paid and unpaid paternity leave and identify predictors of the likelihood of taking paternity leave and the length of leave taken, for both paid and unpaid leave, (2) examine whether and how paternity leave-taking is associated with father involvement, co-parents' relationship quality, and mother's feelings of well-being, and (3) analyze the relationship between paternity leave-taking and children's well-being, and the extent to which father involvement, co-parents' relationship quality, and mother's feelings of well-being mediate this relationship. We examine the patterns and consequences of whether fathers take paternity leave as well as length of paternity leave. Indicators of child well-being include overall health, problem behavior, and cognitive ability. We employ a variety of statistical techniques including regression modeling, propensity score matching, and multilevel models. Overall, a focus on the patterns, predictors, and consequences of paternity leave-taking will provide insights into the potential of paternity leave policies to reduce work-family conflict and promote family well-being. Such insight will inform scholars and policymakers about the implications of the current structure of paternity leave in the U.S., and whether an expansion of these policies may be beneficial to families.
{ "pile_set_name": "NIH ExPorter" }
The NHLBI Electron Microscopy Core Facility has supported projects using the following techniques in the past year: 1. Fixation, embedding, ultra-thin sectioning and transmission EM digital imaging of tissues and cells. 2. EM immunocytochemistry, including immunogold localization of proteins and other antigens within and on the surface of cells by pre-embedding techniques. 3. Negative staining of large proteins, polymers and supramolecular structures as well as lipid and membrane vesicles for transmission EM imaging. 4. Fixation, critical point drying, sputter-coating and scanning EM digital imaging of small organisms, organs, tissues and cells, as well as other materials such as artificial matrices. In the past year, we have supported 26 projects for 17 principal investigators in the NHLBI/DIR: Stewart Levine, James Shelhamer: Autophagy in leukotriene D4-treated mouse macrophage cell line. Robert Kotin: Characterization of AAV preparations for in vivo gene transfer;Course of AAV formation in baculovirus-infected insect cells;Ultrastructure of Duchenne Muscular Dystrophy muscle in dogs. Cecilia Lo: Ultrastructure of cilia in humans with ciliary dyskinesia;Cilia ultrastructure in cardiac development mutants;Developmental defects related to sonic hedgehog signaling in cilia mutant mice. Rosa Puertollano: Regulation of mucolipin-1 induced lysosomal storage granule accumulation in Hela cells by LAPTM4a and 4b; Immunogold localization of novel mucolipin-containing organelles in cultured epithelial cells;Endosome and lysosome regulation by PI-4,5-P-4 Phosphatase Type I. Toren Finkel: Ultrastructural changes resulting from conditional knockout of ATG7 in pancreatic beta cells and skeletal muscle; Mitochondrial morphology in thymocytes of Bmi KO mice. Joel Moss: Lysosomes in liver of ARD1 knockout mice and cell lines. Robert Adelstein: Myocardium ultrastructure in myosin IIB conditional knockout mice. Nico Tjandra: Virus budding in possible internal compartments in transfected Cos cells Claire Waterman: Characterization of patterned fibronectin on acrylamide for cell culture;Characterization of a synthetic surface for cell adhesion studies. Jay Knutson: Characterization of gold nano-particles; Gold nano-particle penetration into tissue slices. Mark Knepper: Validation of urinary exosome assays by electron microscopy;Immunogold localization of aquaporin channel redistribution in cultured epithelial cells. Jennifer Lee: Characterization of liposome-protein interactions. Howard Kruth: Macrophage processing of LDL. Alan Remaley: Micro-RNA binding to HDL. Maurice Burg: Cytoplasmic membrane dynamics during hypertonic stress in Hela cells. Michael Sack: Mitochondrial morphology in Pink 1 knockout mice.
{ "pile_set_name": "NIH ExPorter" }
A Current Regulation by Dipeptidyl Peptidase-Like Proteins Dipeptidyl peptidase like (DPL) proteins, DPP6 and DPP10, regulate Kv4 channel expression and functional properties and are essential components of the native neuronal ISA, somatodendritic A current along with Kv4 alpha subunits and KChIP auxiliary subunits. Without DPL expression, ISA is severely disrupted with reduced current expression and abnormal activation and inactivation properties. In this project we will test the hypothesis that specific conserved functional domains in the first two exons of DPL genes regulate the interaction of DPL proteins with Kv4 channels proteins and determine the functional properties of the channel complex. These studies will provide important new information about the molecular mechanisms that regulate the functional properties of neurons and likely will be important for our understanding of the molecular mechanisms underlying disease processes such as ALS, autism spectrum disorder, asthma, and other regulatory pathways that DPL proteins have been implicated in. In this project we will address the following aims to better understand the molecular mechanisms involved in the regulation of A currents by DPL proteins. Aim 1: Test the hypothesis that DPP6a and DPP10a accelerate inactivation using a novel N-terminal motif that shares a common underlying molecular mechanism with other N-type inactivation domains. Aim 2: Test the Hypothesis that multiple intermediate states are experienced during N-type inactivation by DPP6a and DPP10a. Aim 3: Test the hypothesis that specific DPL residues in transmembrane and peri-transmembrane region modulate Kv4 channel activation gating.
{ "pile_set_name": "NIH ExPorter" }
Real-world experience and clinical research converge in demonstrating that substance-abusers will continue to use drugs despite repeated adverse outcomes on physical and social well-being. This suggests that a characteristic of abuse may be a reduced ability to inhibit dysfunctional action patterns despite clear evidence of their maladaptivity (Paulus et al., 2002). Indeed, a growing body of behavioral, cognitive, electrophysiological and neuroimaging work suggests that cocaine abusers evidence core insensitivity to internally- and externally-generated signals of error. This insensitivity may reduce the ability to guide adaptive behavioral regulation, and may promote increased perseveration on dysfunctional action patterns, including repetitive drug-taking behavior and comorbid criminality. Despite this growing body of work, the nature of these error-monitoring deficits remains largely undifferentiated, however. Error-monitoring is composed of at least two potentially independent processes: an automatic error detection system that monitors for errors in goal-directed behavior and a less automatic system related to the conscious representation of that error. The majority of work evaluating error-monitoring in substance-dependent populations has focused on error-detection, and has reported consistent impairment. Little is currently understood regarding the state of subjective error-awareness in cocaine abusers, however, which precludes firm conclusions regarding the underlying nature of these error-monitoring deficits. Recent developments in electrophysiological and hemodynamic methods now afford the systematic isolation and evaluation of indices of automatic error-detection and conscious error-awareness, respectively. The present proposal seeks to build off these advances by collecting electrophysiological (EEG) and functional magnetic resonance imaging (fMRI) data within a sample of cocaine abusers (and matched controls), to characterize the integrity of neural circuits underlying their automatic and conscious components of error monitoring, respectively. The Mind Research Network has acquired a state-of-the-art mobile MRI scanner, deployed to various male and female correctional facilities, where epidimiological data indicates that over 65% of inmates suffer from serious substance abuse issues. The deployment of this mobile MRI system thus provides unprecedented access to a population of serious cocaine abusers with comorbid externalizing characteristics. With this access, we propose to collect multimodal (EEG/fMRI) data on 138 incarcerated participants, stratified into two groups: those who do (n = 69) and d not (n = 69) meet criteria for cocaine abuse. Participants will perform a time-estimation task that will afford a carefully controlled evaluation of the integrity of both automatic and concious components of error-monitoring within cocaine abusing individuals. In addition, we plan to follow-up our cocaine abusing participants at 1-year post-incarceration, to evaluate the extent to which error-monitoring integrity predicts future abstinence and relapse rates. The data obtained through this project will serve as the most comprehensive investigation of error monitoring abnormalities within a sample of serious cocaine abusers. The ultimate mission of The Mind Research Network is the development of novel treatment programs for serious psychological and mental disorders, and our group envisions the data collected fom this research as an important step towards future work focused on potential therepeutic and treatment applications. PUBLIC HEALTH RELEVANCE: The chronic and repetative nature of drug-taking suggests that substance abusers may have difficulty identifying, and learning from, their maladaptive behavioral patterns. These learning deficits may arise through impairment in the automatic detection of, and/or the conscious awareness of, errors in goal-directed behavior, and may lead to continued drug use. The present study will collect electrophysiological (EEG) and functional magnetic resonance imaging (fMRI) data from a large sample of incarcerated cocaine abusers, to better characterize their error-monitoring deficits, and to investigate the extent to which such deficits predict future abstinence and relapse rates. The results of this work will aid delineation of cognitive and neurocognitive deficiencies underlying substance abuse, and may serve as a gateway for future treatment options that target these cognitive deficiencies.
{ "pile_set_name": "NIH ExPorter" }
This project will test the hypothesis that studying anatomy from 3-dimensional (3-D) computer models enhances learning. Two-dimensional (cross sectional) photographic images of female anatomy from the National Library of Medicine's Visible Human Project will be used to construct 3-D surface contours using a computer program written by the Co-PI running on a Silicon Graphics Workstation. The resulting computer models of anatomical structures will be incorporated into computer aided instructional packages with two levels of flexibility and sophistication. Effectiveness of the different approaches will be tested by first year medical students taking the gross anatomy course who will be assigned to one of three groups using a matched randomized procedure based on initial survey of experience with WWW and Virtual Reality. The same text will be used in each learning package to present the conceptual and factual information, but different modes of visual illustration will be employed. One group will be exposed to traditional static, single-view 2-D labeled illustrations, although these will be derived from reconstructions of the Visible Human data. A control group will study the same text and images printed on paper. The third group will study text linked to 3-D models presented in Virtual Reality Markup Language (VRML) using Netscape and Cosmoplayer. This group will be able to interact with the 3-D models, manipulating their position to allow viewing from virtually any chosen perspective. Both computer packages will be implemented on standard personal computer platforms. To avoid contamination, effectiveness will be measured immediately after training, using a battery of tests including standard multiple choice questions probing understanding of concepts and spatial relationships; identification of structures in cross sectional images; and ability to match the position of a structure with its location within rectangles indicated in otherwise "empty" cross-sectional diagrams. In a quasi-experimental phase, performance of each group on spatial and conceptual questions on the course examinations will be compared. Subjective responses to the experience will be solicited by questionnaire. The subject of these learning packages will be the female pelvis and perineum. These areas of anatomy are particularly difficult to demonstrate by dissection and increasingly important for nearly all physicians to master considering the movement toward generalist medicine, but they are also emotionally charged. This project will have an important impact on health care delivery by improving physicians' understanding and visualization of a difficult and important anatomical area. In addition, issues of instructional design and effectiveness are addressed, including the crucial question regarding cost- effectiveness of using 3-D surface models in education, and platform- independent delivery through the use of hypertext markup language and VRML. The impact of downloading time will be studied to evaluate use via Internet, local area networks, and use from CD-ROM.
{ "pile_set_name": "NIH ExPorter" }
This application focuses on structural alignments at the DNA duplex level associated with abasic sites, exocyclic adducts, 8-oxo-purine lesions and aromatic amine adducts which play a critical role in chemical and radiation induced carcinogenesis. The solution structure of these lesions will be defined by a combination of NMR and molecular dynamics refinements in defined sequence contexts. New directions in our abasic site research will focus on abasic sites containing deletions opposite the lesion and bistrand abasic site lesions which are refractory to repair. These structural studies will be extended to ribonolactone abasic sites both as isolated lesions and as bistrand lesions relevant to neocarcinostatin action. The exocyclic adduct research will be extended to etheno exocyclic deoxycytidine. We shall probe the generality of syn alignments at the exocyclic adduct and the extent of structural perturbations necessary to accommodate the exocyclic ring within the helix. The structural research will be correlated with research on DNA glycosylases which selectively recognize these exocyclic adducts depending on the base opposite the lesion site. We plan a comparative structural investigation of oxidative damage at deoxyguanine and deoxyadenine in the same sequence context in an attempt to understand the origin of the enhanced mutagenicity of 8-oxo-dG in contrast to 8-oxo-dA which is non-mutagenic. An attempt will be also made to characterize imidazole ring-opened FAPY adducts of deoxypurines at the DNA oligomer level. Our previous structural research on base substitution alignments at C8-deoxyguanine aromatic amine adduct sites are being extended to single and double deletion frame-shifts in specific sequence contexts established from in vitro replication studies. Our efforts will attempt to define the conformation of (AA)dG and (AF)dG at the frame-shift site and identify the structural differences associated with single and double deletions. Such a comparative analysis will also be extended to the anti-oxidant carcinogen (ABP)dG and the food toxin (PhIP)dG adducts to evaluate contributions from the aromatic amine ring system to structural alignments at the lesion site. The overall goal is to couple our NMR structural investigations with related calorimetric measurements in Prof. Ken Breslauer's laboratory on these four families of DNA lesions prepared in Prof. Francis Johnson's laboratory. These studies will provide the structural-thermodynamics framework necessary for interpretation of mutagenesis experiments in the laboratories of Profs. Arthur Grollman and John Essigmann.
{ "pile_set_name": "NIH ExPorter" }
DESCRIPTION: The applicant's laboratory has been a forerunner in demonstrating the presence of a H+, K+-ATPase in vascular endothelial and smooth muscle cells. Previous studies had shown the existence of the enzyme in stomach and intestine and renal tubular cells. In vascular smooth muscle cells, the enzyme is responsible for pumping K+ into cells and H+ out, thereby maintaining a relatively high cell pH. This novel ATPase appears to play and important role in agonist-induced contractile response in vascular smooth muscle, and in stimulating nitric oxide production. The enzyme appears to contribute to hypertension in some models, and the hypothesis that inhibition of H,K- ATPase will be efficacious in the treatment of hypertension will be tested. The goal of the present project is to provide a more thorough characterization of the H, K-ATPase system and mechanistic actions in vascular smooth muscle cells and endothelial cells. The project will test the effects of a specific enzyme inhibitor, SCH 28080 (a derivative of imidazo [1,2] pyridine) on renal and cardiovascular function. Both short and long term effects will be evaluated using cultured cells in addition to whole animal studies of rats and dogs. The specific aims are: 1. Quantify the effect of H, K-ATPase on nitric acid (NO) formation in cultured vascular endothelial and smooth muscle cells. Freshly isolated cells will be compared with cultured cells from human umbilical vein and dog coronary artery. In addition to the acute inhibition, aorta will be obtained from rats treated with the H,K-ATPase inhibitor for several weeks. NO will be assayed as NO2 and NO3 metabolites according to the Greiss reaction. 2. Define the relation of cell pH and calcium to NO production during H,K-ATPase inhibition. In some preparations, cell pH will be held constant using the proton- ophore nigericin in combination with acetate n the external media. The possible mediation of ATP and calcium will be tested by measuring ATP, NADPH and Ca2+ concentrations, inhibiting glycolysis (6- phosphofructokinase). 3. Analyze the effect of H,K-ATPase inhibition on vascular smooth muscle pH and contractility (tension) in isolated aorta. 4. Evaluate the effects of H,K-ATPase inhibition of renal function. Clearance and blood flow studies will be performed in anesthetized dogs to assess the effects on glomerular filtration rate (GFR), renal plasma flow, urinary electrolyte and water excretion, and renin release. The possible involvement of NO will be analyzed by observing responses to SCH 28080 before and during administration of L- NAME. 5. Determine blood pressure responses to the H,K-ATPase inhibitor in hypertensive animals (dogs and rats). Arterial pressure will be recorded continuously under conscious, unstressed conditions. The SCH compound will be given acutely and chronically in the before and during inhibition of NO production (with L-NAME). Hypertensive models will include spontaneously hypertensive rat (SHR), angiotensin II infusion hypertension, one and two-kidney models of Goldblatt renal artery-clip hypertension in the rat and angiotensin II in the dog.
{ "pile_set_name": "NIH ExPorter" }
The National Cholesterol Education Program (NCEP) identified diet therapy as the first line of treatment for hypercholesterolemia. NCEP guidelines suggest that a physician and other health professionals implement the Step 1 diet and that a registered dietitian (RD) implement Step 2 diet therapy. In order to evaluate the effectiveness of this approach, the investigators randomly assigned 90 patients (60 males, 30 females) aged 21-65 with hypercholesterolemia and not on hypolipidemic drugs to receive either medical nutrition therapy (MNT) from RDs or usual care (UC) from physicians or nurse practitioners. Dietitians provided MNT using a NCEP based cholesterol lowering protocol which required 2-6 visits over 6 months depending on lipid results. During the 6 month intervention period, physicians and patients agreed not to use cholesterol lowering drugs. Changes from baseline were compared between treatment groups using analysis of covariance controlling for baseline and gender.
{ "pile_set_name": "NIH ExPorter" }
Objective: The goal of the Research Core is to maximize collaboration, leverage investigative strengths, and optimize output of scientific information for reducing metabolic-syndrome related health disparities. The goal will be addressed by a multidisciplinary, translational research approach to the metabolic syndrome and lifestyle change. Specific Aims: 1. Determine the acceptability of and adherence to the Dietary Approaches to Stop Hypertension combination diet (DASH-CD) among African American undergraduate student research volunteers. 2. Determine the impact of the DASH-CD on biomarkers of anti-oxidant capacity and oxidative stress, hemodynarnic, and metabolic variables compared to usual diet alone and with extra minerals and fiber to match the DASH-CD. 3. Assess the relationship of anti-oxidant/oxidative stress biomarkers to the hemodynamic and metabolic variables on the usual diet. Determine the relationship of changes in biomarkers to the other variables with mineral and fiber supplements and with the DASH-CD. 4. Determine the relationships and inter-relationships of behavioral, early development (birth weight), and genetic polymorphisms to expression of the metabolic syndrome and response to nutritional interventions. 5. Assess the impact of high and low-sodium DASH-CD on the above noted variables (Phase 2). Methods: The Research Core will integrate and leverage several freestanding, original, extramuratly funded research projects on the metabolic syndrome ranging from behavioral psychology and clinical physiology to cell and molecular biology into a coherent and unified approach that wilt enhance resource utilization and scientific productivity. The acceptability and effectiveness of the DASH-CD will be defined in an undergraduate young adult (mean age 23) student population of African Americans (97% of student body) and will examine behavioral, early developmental and genetic factors that modulate the expression of the syndrome and responsiveness to lifestyle change. Significance: Obesity and the metabolic syndrome are at epidemic proportions. The genesis of the syndrome is rooted in unhealthy lifestyle patterns established early in life. The Research Core is designed to identify effective lifestyle strategies for improving cardiovascular health in young adults as a prelude to disseminating that information to the community. Moreover, the multidisciplinary, translational research project will provide superb opportunities to train minority investigators with opportunities to support 70 individuals on minority research supplements.
{ "pile_set_name": "NIH ExPorter" }
PROJECT SUMMARY: Many infectious diseases exhibit differential severity between the sexes yet the mechanisms driving such innate biases are incompletely understood. Staphylococcus aureus (SA), including methicillin-resistant (MRSA), is the most common cause of skin and soft tissue infection (SSTI) in the US, and males have a greater than 2-fold higher incidence of infection versus females. Neutrophils are essential for clearance of SA skin infections, and we found that, consistent with murine infection outcomes, neutrophils from female mice are better able to clear SA ex vivo compared to neutrophils from male mice. Major contributors to neutrophil killing of SA include oxidative burst, antimicrobial factors in neutrophil granules, and neutrophil extracellular traps (NET). Therefore, in this exploratory proposal, we aim to align the efficacy of neutrophil phagocytosis and killing of SA ex vivo with measurements of reactive oxygen species (ROS) and nitric oxide (NO) production, NET formation, and changes in gene and protein expression and activation, to define the mechanism(s) of sex-specific phagocyte efficacy against SA. Furthermore, we have identified sex-specific modulators which enhance phagocyte efficacy and will determine the mechanisms by which these modulators augment innate defense. Upon the successful completion of these aims, we will have gained significant insight into the mechanisms of sex-specific phagocyte efficacy against SA, as well as mechanisms and/or targets for augmenting host innate defense.
{ "pile_set_name": "NIH ExPorter" }
For the past several years, this laboratory has been studying the relationship between the ordered environment of the synaptic membrane and G protein mediated signal transduction systems. Of particular interest has been the capability of elements of the cytoskeleton to alter the coupling among receptors and G proteins involved in the stimulation or inhibition of adenylyl cyclase. Given the increasing number of processes attributed to G proteins, some mechanism which channels individual receptors, G proteins and effectors is likely to exist. This might be particularly true in the nervous system, where rapid and discrete response is a hallmark of synaptic transmission. Most recently, it has been observed that a subunits of G proteins may form complexes with synaptic membrane tubulin and undergo a directed transfer of nucleotide from the latter. This appears to be a highly specific process, as tubulin has been shown to bind, with high affinity, to only two G proteins, as and ail. Even though several other G proteins (alphai2, alphai3, alpha o and transducin (alpha r) are quite closely related to alpha il, their affinity for tubulin is much lower. The initial objective of research in this proposal is designed to determine the binding sites on tubulin for ail or as and the binding sites on these G proteins for tubulin. Studies will be done whereby a variety of proteolytic digestion (both tubulin and G proteins) products are subjected to binding of labelled G protein or labelled tubulin. These studies will be done with native and unfolded proteins and pep- tides, and the interaction will be quantified. Studies with chimeric G proteins, which contain portions of high- and low-tubulin affinity G alphas,will also be performed in the hope of identifying accessible tubulin binding regions. Since different regions on the tubulin molecule appear to be involved in G protein binding and nucleotide transfer, attempts to identify both regions are planned. Reconstitution studies with purified components will be employed to see whether other constituents of the synaptic membrane, which are known to interact with tubulin, might influence the complex formation, or the transfer of nucleotide, between tubulin and G proteins. Finally, peptides corresponding to the tubulin or G protein "binding sites' and "nucleotide transfer domains' will be synthesized. After a thorough characterization of these peptides in reconstituted systems, those which disrupt tubulin-G protein interaction will be tested for their effects on G protein-mediated processes in neural membranes and cells. It is hoped that these studies will provide insight as to how the cytoarchitecture of the neuron contributes to neuroreceptor response and responsiveness. Increase in the understanding of synaptic function must lead to increased understanding of the inner working of the brain.
{ "pile_set_name": "NIH ExPorter" }
DESCRIPTION Abstract Certain inherited modes of heart failure and several neurodegenerative diseases are characterized by protein misfolding states whose underlying mechanism(s) and pathophysiology are poorly understood. Effective therapies exist primarily as goals, not as clinical implementations. Macromolecular damage induced by oxidative stress is the sine qua non for thinking about many diseases. Our laboratory has challenged this paradigm by demonstrating that mouse hearts exhibiting protein-folding cardiomyopathy found in humans are under 'reductive stress' from an over-active antioxidative system. Decreasing the function of glucose-6-phosphate dehydrogenase (G6PD), which generates the reductant NADPH, cures the disease in mice by ameliorating reductive stress, aggresome formation, hypertrophy, heart failure and death. This experiment defines a novel causal mechanism and implicates G6PD as a potential therapeutic target. We hypothesize that stress response and anti-oxidative pathways undergo a pathogenic transition, and become dysregulated by macromolecular stresses (e.g., misfolded proteins). We further propose that other cardiac and neurodegenerative diseases result from similar pathogenic transition. Our Pioneer Award proposal is designed to develop a robust experimental platform for exploring the mechanisms of reductive stress disease. Our work will extend from studies that model reductive stress in the genetically amenable fruit fly Drosophila melanogaster, through cultured mouse and human cells, to whole mice, and finally into patients as we work to develop diagnostic tools. Cuttingedge imaging techniques will be developed for monitoring redox couples and toxicities in living cells and tissues. Genetic screens in Drosophila will guide the identification of new genes and determine the effects of potentially therapeutic compounds that prevent reductive stress disease. Validation in mice of interacting genes and pathways will provide us target candidates for pharmacolog
{ "pile_set_name": "NIH ExPorter" }
The proposed research will examine in detail the role of poly (ADP-ribose) in DNA damage and DNA repair in human fibroblasts from normal, xeroderma pigmentosum patients, and parental heterozygotes. DNA damage will be induced by treatment with N-methyl-N'-nitro-N-nitrosoguanidine and UV-irradiation. A newly developed sensitive and selective chemical assay will be applied to measure the total intracellular levels of poly (ADP-ribose) following DNA damage. The size distribution of polymer chains and the intracellular levels of the precursor NAD will also be studied in order to estimate the rate of turnover of poly (ADP-ribose). The metabolism of poly (ADP-ribose) in vivo will be related to the activities of poly (ADP-ribose) polymerase and poly (ADP-ribose) glycohydrolase which will be measured independently in nucleotide permeable cells. All of the parameters of poly (ADP-ribose) metabolism will be quantitatively related to the amount of DNA damage using velocity sedimentation on alkaline sucrose density gradients and to the amount of unscheduled DNA synthesis using equilibrium sedimentation on alkaline cesium chloride density gradients. Since poly (ADP-ribose) is formed from NAD which is derived from the vitamin niacin, the results will be carefully examined for potentially significant nutritional factors which relate to environmental carcinogenesis.
{ "pile_set_name": "NIH ExPorter" }
DESCRIPTION: The broad objectives of the parent proposal for this FIRCA are to understand microtubule dynamics in vivo and in vitro and, in particular, to determine the nature of the coupling between chromosome movement and microtubule growth and shortening. Specific aims targeted in the current grant period focus on analysis of microtubule formation, disassembly, turnover, and selective stabilization. Cell systems to be investigated include mammalian fibroblasts, mitotic cells, and neurons. This proposal addresses an apparent paradox in the MT dynamics field. Although dynamic instability is now established as a primary mechanism of MT turnover, consideration of the totality of evidence in vitro and in vivo suggests that dynamic instability, as originally formulated, cannot account for the rapidity of turnover in vivo. The specific hypothesis to be tested in this proposal is the "conveyor assembly" model for centrosome activity first proposed by the foreign collaborator in 1983. According to this view, the MT array is developed by initiation of MTs at the centrosome followed by their detachment (release) and centripetal displacement away from the centrosome. Two key predictions of the conveyor assembly model will be tested: (i) minus ends of MTs in cultured cells are rather stable; and (ii) MTs are continuously released from the centrosome.
{ "pile_set_name": "NIH ExPorter" }
This subproject is one of many research subprojects utilizing the resources provided by a Center grant funded by NIH/NCRR. The subproject and investigator (PI) may have received primary funding from another NIH source, and thus could be represented in other CRISP entries. The institution listed is for the Center, which is not necessarily the institution for the investigator. Arthritis is the leading cause of disability in the United States in persons eighteen years and older. Disability arising from arthritic conditions accounts for tremendous health care costs for individuals, their families, employers, and the country. In fact, the Center for Disease Control estimated arthritis-related medical costs are $15 billion annually. Currently available therapies are often ineffective in controlling autoimmune inflammatory arthritis like rheumatoid arthritis (RA) and psoriatic arthritis (PsA). Understanding the mechanisms that cause and regulate arthritis is critical for the development of new, better and more specific therapies. Both rheumatoid arthritis and psoriatic arthritis synovial tissues have a highly invasive behavior that leads to cartilage and bone destruction. The purpose of the synovial tissue collection program is to obtain human synovial tissue samples from patients with rheumatoid arthritis, psoriatic arthritis and controls (normals undergoing trauma related surgery, or osteoarthritis). These tissues are essential for the identification of differentially expressed genes that may account for the disease-associated invasive and destructive behavior. Additionally, primary synovial cell lines will be generated from these tissues, and used to validate the gene expression findings from tissues, and well as in in vitro functional studies. It is anticipated that these studies will identify novel therapeutic targets. Assays that will be performed using this material include biochemical assays to determine the activation and invasive status, and microarray experiments. Quantitative Reverse Transcriptase Polymerase Chain Reaction and immunohistochemistry will be used to validate the microarray findings.
{ "pile_set_name": "NIH ExPorter" }
This revised proposal describes a 5-year program for the development of the candidate's career as an independent clinician-scientist focused on lung vascular biology.,The candidate, an Assistant Professor in UCSF's Department of Surgery with research experience in the cardiopulmonary system, will perform this work with strong institutional support in an environment that includes an outstanding research program in vascular biology. I propose to study the role of Ndtch4 signaling in the development of lung arteriovenous malformations (AVMs). Mice that constitutively express endothelial Notch4 develop AVMs in extrapulmonary sites as well as hemorrhages in the lungs and filling defects on pulmonary angiograms. Common bile duct ligation induces lung AVMs in rats and impairs lung function in mice. I hypothesize that Notch4 signaling is necessary and sufficient for the development of lung AVMs. The specific aims are: 1) To determine the effect of constitutively active endothelial Notch4 on the structure and function of the mouse lung vasculature. 2) To evaluate lung vascular remodeling after common bile duct ligation in the absence of Notch4. 3) To determine the effect of lung-AVM-patient serum on human pulmonary endothelial cells. The experiments in this proposal will use vascular casting, microcomputed tomography, and other indirect methods to rigorously characterize the lung vasculature in the context of Notch4 signaling. First, mouse lungs will be evaluated after different durations of constitutive endothelial Notch4 expression. Second, the mouse common bile duct ligation model will be further characterized and the necessity of Notch4 for lung vascular changes will be tested by performing the procedure on Notch4-/- mice. The final aim will establish the relevance of aims 1 and 2 in the human disease process and will provide evidence for a "hepatic factor" that regulates lung vascular remodeling. Specifically, cultured human pulmonary endothelial cells will be exposed to serum from patients with documented lung AVMs, and both Notch signaling and endothelial cell behavior will be assessed. This work will determine the role of Notch4 expression in the development of lung vascular abnormalities and will begin to characterize the cellular and molecular mechanisms involved. RELEVANCE: Lung arteriovenous malformations are a significant clinical problem for which there is no effective medical treatment. The ultimate goal of this research is to lead to new molecular targets of therapy for patients with lung AVMs and other pulmonary vascular disorders.
{ "pile_set_name": "NIH ExPorter" }
In this project, we propose to continue our investigation of the biochemical and molecular mechanism(s) by which "insulin resistance" is acquired. Our long-term focus has been on the potential link between nutrient excess and impairment of insulin action. In this regard, we have recently shown that increased nutrient (eg, carbohydrate and lipid) availability results in insulin resistance and in increased leptin gene expression via increased flux of carbons into the hexosamine biosynthetic pathway. We propose herein that a close loop feed-back regulation is normally operating between nutrients and their metabolic outcomes. Thus, nutrient excess is sensed via the hexosamine biosynthetic pathway and generates signals leading to decreased insulin action on glucose uptake and to increased leptin gene expression. They also favor increased storage into lipid via increased tissue levels of Malonyl-CoA and Long Chain-CoA and ultimately increased adiposity and weight gain. However, the concomitant induction of leptin expression attempts to counteract this drive by antagonizing the effects of nutrients on Malonyl-CoA and triglyceride storage, on the hexosamine pathway and perhaps via direct effects on insulin signaling. Any disruption of this physiological response (due to either impaired stimulation of leptin expression by nutrients or to decreased effectiveness of leptin action on target tissues) is likely to lead to increased adiposity and more insulin resistance. Based on preliminary results and on this overall hypothesis we wish to pursue the following specific aims: 1. How do nutrients regulate insulin action? We will focus on the interaction between increased lipid availability and skeletal muscle insulin signaling and action. We hypothesize that the susceptibility to develop insulin resistance in response to excessive nutrient exposure is modulated by the skeletal muscle ability to utilize fructose-6-phosphate in the glycolytic pathway. 2. How does leptin modulate hepatic and muscle glucose/lipid metabolism and insulin action? We will examine whether specific hypothalamic targets of leptin play distinct roles in mediating its complex metabolic effects. 3. Is leptin synthesis and/or action modulated following prolonged stimulation? We will generate short-term models of relative hyperleptinemia and will test the hypothesis that the "protective" effect of leptin against nutrient excess wanes if the leptin signal/transduction system is chronically over-stimulated.
{ "pile_set_name": "NIH ExPorter" }
Addition of an oral hypoglycemic agent to a regimen including insulin treatment of non-insulin dependent diabetes results in a decrease in the dose of insulin required for optimal glycemic control. The hypothesis that this decrease in exogenous insulin administration will result in improvement in the degree of obesity, a decrease in blood pressure, and a decrease in blood lipids, independent of glycemic control, will be tested using a double blinded, placebo controlled, crossover design with a washout period between treatments.
{ "pile_set_name": "NIH ExPorter" }
HIV enters host cells through several steps, including the formation of an entry complex, the fusion process, and migration toward nucleus for viral replication. The viral envelop gp41-gp120 heterodimers associate into gp41/gp120 trimers to form spikes on the viral surface. Structural studies suggest that a mature HIV particle contains 10-15 spikes each forming a knob with a diameter of 10.5 nm. Binding of CD4 to gp120 leads to conformational changes in a spike that allows gp120 to interact with a chemokine receptor, which completes the formation of an entry complex. Ligation of gp120 to CD4 and a chemokine receptor triggers further structural changes that allow gp41 to insert into the target cell membrane, which triggers the fusion between viral envelop and host cell membrane. Upon the fusion, vesicles that contain virus migrate through cortical actin barrier towards nucleus. In macrophages or T cells where re-organization of actin cytoskeleton is spontaneous and robust, the cortical actin presents little resistance for the HIV migration. Therefore, it appears that chemokine receptor signaling that leads to actin de-polymerization is not essential for HIV infection. In resting T cells, however, it has been demonstrated that CXCR4 signaling mediated de-polymerization of cortical actin matrix is essential for HIV replication. We will image pseudo HIV virus-induced reorganization of cortical actin in live cells using the same imaging setup. Pseudo viruses labeled by Cy5 will be placed on the surface of the cover glass, and resting CD4 T cells will be placed on top of the viruses. Using time-lapse experiments, we will capture the virus-triggered changes of cortical actin during HIV entry process in real time. To test whether a cell can prevent HIV entry by stabilizing its cortical actin, we will first treat cells with jasplakinolide, an F-actin stabilizing agent, and then track HIV viruses using time-lapse movies. The movement of viruses will be analyzed by our developed software package to obtain parameters, such as diffusion constants and types of the movement. Such dynamic information is crucial for a more complete understanding of HIV entry and mechanism of reorganization of cortical actin.
{ "pile_set_name": "NIH ExPorter" }
Recent evidence has clearly shown that cytokines can suppress experimental allergic encephalomyelitis (EAE), a rodent model of central nervous system (CNS) autoimmune disease. These cytokines influence the activation and/or expansion of the Th1 effector subset of CD4+ T cells which not only induce EAE but are the effectors of a number of autoimmune diseases. This proposal is based on differential CD4+ responses induced in EAE susceptible female (Th1 responsiveness) and EAE resistant male SJL mice (Th2 responsiveness). This dichotomy of female susceptibility is consistent with the increased prevalence of a number of human autoimmune diseases including multiple sclerosis, myasthenia gravis, rheumatoid arthritis and SLE in females. Thus, the sex-based difference in CD4+ T cell responsiveness in SJL mice provides the unique opportunity to explore induction and suppression of Th1 and Th2 cells in an in vivo model using a single mouse strain. Adoptive transfer of a mixture of myelin basic protein-specific Th1 effector/Th2 suppressor cells into EAE susceptible mice has a dramatic suppressive effect on EAE compared to recipients of Th1 effectors only. This proposal determines both the constraints imposed on Th2-mediated EAE suppression and if Th2 cells must access the target organ to suppress disease. Another novel aspect of this model is that Th2 responses in EAE resistant male mice are reversed to Th1 responses via adoptive transfer of an antigen presenting cell (APC) subset derived from EAE susceptible female mice. The mechanism of APC-dependent CD4+ T cell polarization is examined by monoclonal antibody-mediated ablation of Th2 cytokines (IL-4 and IL-10) and increases in Th1 cytokines (IL-12 and IFN-gamma) using recombinant cytokine. A direct effect on APC-dependent priming versus expansion of EAE effectors will be determined by examining phenotype changes in APC derived from cytokine treated males exhibiting the EAE susceptible phenotype. This proposal explores the basis for APC-dependent in vivo regulation of Th1/Th2 priming to self antigens, which plays a determinant role in the outcome of a number of autoimmune diseases. Information on the induction and regulation of Th1/Th2 cells may provide a viable alternative for cytokine-based immunotherapies and suggest the possibility that induction or transfer of autologous Th2 cells may be useful in ameliorating autoimmune disease.
{ "pile_set_name": "NIH ExPorter" }
Patients with ischemic left ventricular dysfunction whoa re suspected of having stunned or hibernating myocardium will be enrolled in this study to evaluate the safety and efficacy of D-ribose infusion for improving myocardial wall motion abnormalities detected by echocardiography.
{ "pile_set_name": "NIH ExPorter" }
An onging dissemination effort organized by Resource Assistant Director Dr. Kevin Yarasheski is the establishment of an annual symposium on application of stable isotope tracer methods and mass spectrometry to the study of human biology. The first such symposium was held at the Washington University Medical Center on July 18, 1998. The symposium was chaired by Dr. Kevin Yarasheski, Assistant Director of the Medicine Department site of the Mass Spectrometry Resource, and it was entitled "Bridging the gap between ion chemistry and biomedical applications. 15N/14N Analysis bi gas chromatography-combustion-reduction-isotope ratio mass spectrometry." The Conference sponsors were the Washington University Mass Spectrometry Resource, Cambridge Isotope Laboratories, Finnigan MAT, MassTrace, Inc., and the Washington University Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism. Scientists from around the nation and Europe attended the conference. The conferen ce objectives were to inform scientists about the utility, precision, and accuracy of GC-combustion-reduction-IRMS for determination of 15N/14N isotope abundance in biological compounds and to educate scientists about the biomedical questions now being addressed with this technology. Conference speakers and their topics are listed below in Table 5. Next year's annual conference will focus on lipid metabolism and its characterization by isotope ratio mass spectrometry. This conference will be held as a satellite symposium of the 1999 meeting of the American Society for Mass Spectrometry (Dallas, TX, June 13-18, 1999) or of the 1999 Experimental Biology meeting (Washington DC, April 17-21, 1999).
{ "pile_set_name": "NIH ExPorter" }
The applicant's long-term objectives are to define the biochemical and genetic basis for human inherited neuromuscular disorders. In line with his previous work on Duchenne and Becker muscular dystrophies, the applicant proposes to apply molecular genetic, biochemical and cell biological techniques towards this goal, using diagnostic muscle biopsies, DNA, and clinical data as his primary materials. In this proposal, the applicant describes plans to continue to develop dystrophin testing as a means for biochemical diagnosis of myopathies. Dystrophin testing appears capable of detecting all Duchenne/Becker dystrophy patients even before the onset of obvious clinical symptoms, and the results are clearly prognostic. DNA testing, on the other hand, is only capable of detecting two-thirds of Duchenne/Becker patients, and the results are much less prognostic of the expected clinical severity. The proposed biochemical studies will delineate the range of clinical phenotypes possible from dystrophin abnormalities by investigating dystrophin abnormalities in limb-girdle and congenital myopathy patients. An important result of the continued patient testing will be the establishment of an extensive patient database of true limb-girdle and congenital myopathy patients. This database will be used as starting material for investigations directed towards identifying underlying biochemical defects in patients not having dystrophin abnormalities. The experimental approach to be used for identifying the protein defects in these patients will be the candidate gene/protein approach. Specifically, we have recently developed experimental methods by which we can clone human genes and produce large quantities of the corresponding human protein from any published vertebrate sequence with only a few hours of work. Antibodies raised against the produced proteins have proven to highly sensitive and specifically directed against the human protein. In this proposal we describe our application of these techniques towards two human proteins, the chromosome-6 dystrophin-related protein, and the skeletal muscle sodium channel. The cloned human genes and the corresponding antibodies will be used to investigate the possible involvement of these proteins in neuromuscular disease.
{ "pile_set_name": "NIH ExPorter" }
The sarcolemma (plasma membrane) of myocardial cells has high levels of Na+-Ca2+ exchange activity. Na+-Ca 2+ exchange is an important mechanism for regulating myocardial Ca 2+ and thereby also regulates contractility. This laboratory has recently cloned the Na+-Ca 2+ exchanger allowing molecular analysis of the exchanger to begin. The long-term objective of this project is to understand the distribution, regulation, and gene structure of the exchanger. Towards this goal, the specific aims of the project are as follows: 1. Isoforms and distribution. cDNA clones representing isoforms of the Na+-Ca2+ exchanger will be isolated, sequenced and expressed. Isoforms function and distribution will be assessed. 2. Regulation. The effects of thyroid hormone, chronic digitalis treatment, and ischemia on exchanger protein and RNA levels will be determined. These three interventions all affect Na+-Ca2+ exchange activity, and the proposed experiments will analyze these effects at the molecular level. 3. Gene structure. Transcriptional regulatory elements and intron/exon boundaries will be analyzed. Characterization of upstream regulatory elements will aid in understanding the regulated expression of different exchanger gene products. Intron/exon maps are useful in understanding evolutionary and protein domain relationships. 4. Antisense oligonucleotides. Oligonucleotides with sequences antisense to mRNA for the Na+-Ca2+ exchanger will be introduced into cells. This should block synthesis of exchanger protein and the response of excitation-contraction coupling pathways will be analyzed. The Na+-Ca 2+ exchanger is an important regulator of contractility. Further molecular knowledge of this essential cardiac transporter will aid in understanding the workings of both normal and diseased hearts.
{ "pile_set_name": "NIH ExPorter" }
Adapatations of the Temporomandibular Joint Disk NO ABSTRACT ON FILE
{ "pile_set_name": "NIH ExPorter" }
Surveillance data from large and small spatial scales play an essential role in public health decision making and the scientific investigation of infectious disease. However, such data are subject to missing observations, delays in reporting, and observational biases that can lead to increased uncertainty and incorrect conclusions. Existing uncertainty and bias are amplified when we attempt to predict future disease incidence. This project aims to develop and extend statistical and modeling methodologies to correct for biases in surveillance data, impute missing data, predict the course of epidemics, and appropriately characterize uncertainty at relevant spatial scales. An integrated Bayesian framework and Markov chain Monte Carlo methods will be used to combine data from multiple sources at different spatial scales to better understand macro- and micro-scale dengue dynamics in Thailand. Dengue is a mosquito-borne virus which circulates in over 100 countries, is reemerging throughout much of the western hemisphere, and is responsible for an estimated 50 million infections and 19,000 deaths worldwide each year. Specifically, this projects aims to: (1) develop an integrated statistical framework for predicting missing surveillance data by using information from correlated locations and non-traditional data (e.g., search queries); (2) develop methods that exploit mechanistic models of disease transmission and spatial smoothing techniques to bridge the gap between data collected at differing spatial scales while appropriately quantifying uncertainty; and (3) create improved methods for the analysis of spatio- temporal point pattern data that: appropriately account for biases in data collection, elucidate patterns of spatio-temporal dependence at the appropriate scale and define the spatial scale of disease transmission. Methods will be tested and validated using simulated data sets, over four decades of province level dengue surveillance data collected by the Thai Ministry of Public Health, district level surveillance level for over 20 years from several Thai provinces, and point pattern data on the exact location where dengue cases presenting at select hospitals reside. This project will result in the creation and dissemination of novel methods for dealing with bias, missing data and uncertainty in regional, local and point pattern statistics. These methods will aid the appropriate interpretation of macro-levels statistics at the local level and create new tools for using point pattern data to answer scientific questions about disease spread. In addition, fulfillment of these aims will increase our understanding of dengue transmission in Thailand at a variety of spatial scales, improve the Thai dengue surveillance system by providing an integrated approach for predicting future incidence and imputing missing data, and aid in modeling and responding to emerging dengue epidemics elsewhere in the world. The resulting methods will be disseminated in peer reviewed publications and R packages (including source code and validation data sets) freely available on the Comprehensive R network; allowing public health practitioners and researchers from a variety of disciplines to utilize and build upon this work.
{ "pile_set_name": "NIH ExPorter" }
ABSTRACT Many bacteria are motile by synthesizing corkscrew-like flagella which when rotated propel bacteria through the environment. Each bacterium synthesizes a species-specific number of flagella and inserts the flagella in a species-specific pattern on the cell surface. Flagella are complex nanomachines assembled from dozens of different proteins and how each bacterial species controls flagellar number and patterning is poorly- understood. Moreover, the number of flagella per cell increases when cells come into contact a solid surface to initiate a form of surface motility called swarming. The Kearns lab uses classical forward genetics, super- resolution microscopy, and biochemistry to study flagellar biosynthesis and swarming motility of the Gram positive bacterium Bacillus subtilis. The goals of the project are to understand flagellar biosynthesis in the context of growing cell architecture. First, we will determine how flagellar number is controlled by the poorly- understood master regulator of flagellar biosynthesis SwrA and a response regulator DegU. Second, we will explore how the surface contact response is transduced to inhibit the adaptor-mediated regulatory proteolysis of SwrA and increase flagellar number. Third, we will learn how the flagellar rod insertion through peptidoglycan occurs, and how rod length is controlled to match the thickness of peptidoglycan. Fourth, flagella are synthesized in a grid-like pattern and we will study how flagellar patterning is interpreted and updated in time during cell growth, and coordinated with peptidoglycan insertion. Ultimately, we want to achieve a holistic understanding of how a cell dynamically governs the initiation of flagellar biosynthesis at specific locations to insert the machine through the cell envelope. Our basic research is fundamental to how cells self-organize and is applicable to the spatiotemporal control of the assembly of transenvelope nanomachines involved in pathogenesis including flagella, pili and secretion systems like the injectisome.
{ "pile_set_name": "NIH ExPorter" }
Wehavedevelopedanalgorithmforcarryingoutquantumchemical calculations in a dielectric medium. Our electronic structure program PSGVB first calculates electrostatic point charges using an ESP fitting method. These charges are then passed to DELPHI which determines the reaction filed by solving the Poisson-Boltzmann equation. The reaction field is represented as point charges on the dielectric boundary. These are passed to PSGVB which now solves quantum chemical equations in the field of the point charges. This process is iterated until self-consistency is reached. We have shown that if one uses accurate gas phase quantum chemical methods (GVB plus a good basis set is sufficient) solvation energies accurate to ~0.6kcal/mole can be computed for a test set of 29 molecules. Recent work includes development of an analytical gradient method using a novel finite element Poisson-Boltzmann solver and improvement of the accuracy of the continuum method via inclusion of corrections for short-range hydrogen bonding terms.
{ "pile_set_name": "NIH ExPorter" }
Boucher, Richard C. CORE A: PROGRAIUI MANAGEMENT CORE Investigators P.I.: Richard C. Boucher, Professor of Medicine Co-Investigator: Claire Doerschuk, Professor of Medicine A. DESCRIPTION OF THE CORE UNIT The Program Management Core will essentially function as a drug development decision-making body. Thus, it will oversee all aspects of the drug development paradigm, and program administration. Overall program management will be the responsibility of the P.I., assisted by the Co-I., in coordination with three advisory groups (see Figure 18, Program Introduction): 1. Project P.l.s and Core P.l.s; 2. Internal Drug Development Advisory Committee, including: a. Dr. Stephen Frye, Director of the Medicinal Chemistry and Novel Molecular Entity Screening Core, UNC CTSA/School of Pharmacy. Note, Dr. Frye is a former worid-wide head of medicinal chemistry for GSK and has been at UNC as Director of the CTSA MedChem Core for the past three years. b. Dr. Anthony Hickey, Ph.D., Professor of Pharmacy, UNC School of Pharmacy. Dr. Hickey is an expert in aerosol drug delivery, and the founder of Cirrus Aerosol Development Corp. and Oriel Inhaled Device Company. c. Dr. Lisa Lavange, Professor in Practice, Director, Carolina Computing Center. Dr. Lavange is a former VP for Biostatistics at Inspire Pharmaceuticals and Senior Statistician at Quintiles. d. Dr. Joseph Eron, Professor of Medicine. Dr. Eron is an experienced clinical trials expert in HIV and serves on the UNC Conflict of Interest Committee. 3. External Advisory Group - TBN Central to all aspects of the direction/oversight of the Program Oversight Committee is the integration of advice from the Project and Core P.l.s, the Internal Drug Development Committee, and the External Advisory Board into the decision-making process. To accomplish this goal, there will be monthly meetings of the tPPG P.I./co-P.I. with the Project and Core P.l.s and the Internal Drug Development Committee. In addition, there will be quarteriy teleconferences (one meeting/yr will be face to face) with this group with the External Advisory Committee to discuss progress and seek input. It is likely that the External Advisory Committee will consist of three individuals with the following skills: 1) expertise in mucus biochemistry/biophysics; 2) industry drug development; and 3) clinical trials in COPD. Finally, as part of this overall program management function, the requisite administration,
{ "pile_set_name": "NIH ExPorter" }
It is critical to the future of pediatric cardiology that talented clinician-scientists continue to be fostered and encouraged to conduct research related to childhood cardiovascular disorders. This proposal requests a continuation of the University of Iowa's pediatric cardiology Institutional Ruth L. Kirschstein NRSA, established in 1978. The goal of this proposal is to foster multidisciplinary research training for three MD and MD/PhD postdoctoral trainees per year in clinical, basic science or population research. The two- to three-year training program will serve as a foundation for a career in academic pediatric cardiology. A diverse group of mentors from clinical, basic science, public health and engineering departments use state-of-the-art techniques to explore specific questions in genetics, molecular and cellular biology, physiology, cardiovascular imaging, and epidemiology. The co-Program Directors and the Internal Executive Committee of the Training Program guides trainees through the early stages of selecting a mentor, or mentor team, and choosing a research discipline and monitors their progress during training. Trainees attend structured lectures and group discussions on many of the aspects needed for successful academic careers. Attention is given to career development with time available for interaction with other scientists at local and national meetings, and manuscript and grant preparation. Since its inception, our program has trained 46 pediatric cardiologists to study diverse topics from cardiac development and ontogenesis to the prevention of childhood obesity. After completing this program, nearly 85% took academic appointments and a significant number (46%) continued their research interests by obtaining NIH funding (N=21). This proposal requests ongoing support of our program, which 1) recruits talented applicants who desire a career in academic pediatric cardiology with an emphasis on research and 2) fulfills the recommendation of the NIH Task Force to encourage and support clinicians interested in becoming independent investigators of pediatric cardiovascular diseases. Relevance: Congenital heart disease is the most common birth defect. Increasing our understanding of the origins and treatments of cardiac abnormalities children and adolescents requires that we continue to train talented clinician scientists in pediatric cardiology. The goal of this proposal is to continue funding for program with a long track record of training researchers in pediatric cardiology.
{ "pile_set_name": "NIH ExPorter" }
The suppressor of cytokine signaling (SOCS)- genes, SOCS1 and SOCS3, are critical targets of signal transducer and activator of transcription (STAT) pathways, providing normal negative feed-back control of signaling by cytokines and growth factors. Consistent with STAT activation in many tumor cells, SOCS1 expression is silenced in human cancer and enforced SOCS1 expression inhibits tumorigenesis. SOCS3 is closely related to SOCS1, but its regulation and role in oncogenesis remain largely unknown. Our preliminary studies find that cells transformed by Lck exhibit active STATS but fail to express both SOCS genes. In contrast to SOCS1 silencing by DNA methylation, defect of SOCS3 expression may be linked to the absence of STAT5b phosphorylation on non-conserved serine sites identified by our mass spectrometry. Additional preliminary studies show that SOCS3 associates with Lck in cells and becomes tyrosine-phosphorylated, and ectopic SOCS3 expression results in short-term growth suppression of Lck-transformed cells. Our long-term objectives are to determine how expression of different SOCS genes is disrupted in malignancies and to gain new mechanistic insights of their roles as tumor suppressors in various tumor models. This proposal focuses on the regulation of SOCS3 gene expression and protein functions. Aim 1 will determine the role of novel STAT5b serine phosphorylation in SOCS3 expression. We propose to define the functional STAT5b serine phosphorylation sites and elucidate its role in STAT5b-dependent assembly of transcriptional complex on the SOCS3 promoter in the context of chromatin. Aim 2 will test our hypothesis that SOCS3 and Lck proteins mutually regulate each other's functions and characterize the underlying mechanism. We further propose that a tyrosine mutant of SOCS3 is more potent than wild-type SOCS3 in suppressing Lck-mediated oncogenesis. In Aim 3, different SOCS3 proteins will be expressed in Lck-transformed LSTRA cells to evaluate their tumor-suppressing activities both in culture and in mice. The possibility to restore endogenous SOCS3 expression will also be tested with potential gain-of-function STAT5b mutants in LSTRA. Using Lck as a model system, we will start to define molecular details underlying the differences between SOCS1 and SOCS3 in their gene regulation and protein functions. Functional characterization of novel serine phosphorylation sites in STAT5b will further establish a new paradigm for its role in regulating STAT signaling and STAT-mediated gene expression. Relevance: SOCS proteins are normally produced by cells to turn off signals that stimulate their growth. Many cancer cells lose expression of these potential tumor suppressors. Studies proposed here will help us better understand why SOCS genes are not expressed in human cancer and how SOCS proteins may function differently in tumor cells. Collectively they will reveal potentially new targets for cancer therapy. [unreadable] [unreadable] [unreadable] [unreadable] [unreadable]
{ "pile_set_name": "NIH ExPorter" }
Malaria and other human diseases caused by mosquito-borne pathogens are major threats to global health. The spread of insecticide-resistant mosquitoes, drug-resistant parasites and the lack of effective or affordable vaccines have all led to an alarming increase in disease burden over the past few decades. A vital component of any comprehensive mosquito-borne disease prevention strategy is to gain a better understanding of the physiology of mosquitoes. I will investigate fundamental questions of mosquito biology involving the regulation of juvenile hormone synthesis. This hormone plays a central role in coordinating metamorphosis and the female reproductive cycle. Interfering with the regulation of juvenile hormone production has potential as a safe and effective vector control strategy. During a sabbatical year at Florida International University, in the laboratory of Dr. Fernando Noriega, I will learn methods and approaches to investigating intracellular signaling pathways in the Yellow Fever mosquito, a commonly used model system for mosquito endocrinology. Dr. Noriega has an NIH R01 supported research program that is investigating the regulation of juvenile hormone production in mosquitoes. I am particularly interested in investigating the signaling pathways that allow peptide hormones to either increase or decrease the production of juvenile hormone by a pair of endocrine glands called the corpora allata. PUBLIC HEALTH RELEVANCE: This research experience will significantly enhance my mosquito research at Muhlenberg College and allow me to change the focus of my current research program from the molecular to the cellular level.
{ "pile_set_name": "NIH ExPorter" }
This is a resubmission of 1 K07 CA112529-01 "Hormones, hormone-related genes, and colorectal cancer." Candidate: Jennifer Hsiang-Ling Lin received her Ph.D. in 1997 at the University of Missouri-Columbia. She subsequently worked as a biostatistician in the Division of Biostatistics, Washington University in St. Louis between 1997 and 1999, and later began her research work in the field of statistical genetics in the Psychiatry Department at Washington University in St. Louis between 1999 and 2002. She is currently an Instructor in Medicine at Harvard Medical School (HMS). She is applying for this Career Development Award to acquire the methodological and theoretical research skills needed to become an independent scientist in cancer genetics and epidemiology. Sponsor and Environment: l-Min Lee, MBBS, ScD, is an Associate Professor of Medicine at HMS and co-investigator of the Women's Health Study (WHS). Nancy R. Cook, ScD, is an Associate Professor of Medicine (Biostatistics) at HMS and also co-investigator of the WHS. They have extensive experience training investigators in the fields of lifestyle and chronic diseases as well as numerous publications in these areas. Research: The proposed study will examine the associations between exogenous hormone use, reproductive factors, estrogen-synthesizing genes, estrogen and progesterone receptors, and colorectal cancer (CRC) risk in a large cohort of women from the WHS, a randomized clinical trial of vitamin E and low-dose aspirin in the primary prevention of cancer and cardiovascular disease among 39,876 women. As of 2007, an estimated 413 CRC cases will have occurred in the overall WHS cohort (288 with blood specimens). This proposed study will provide detailed assessment of the long-term effects of postmenopausal hormone therapy on CRC risk according to duration, formulation, and dosage in the WHS female cohort. In addition, the study will investigate duration of oral contraceptives use, parity, and cumulative exposure to endogenous estrogen (estimated from duration between menarche and menopause) in relation to CRC risk. Finally, the study will comprehensively examine the associations between putative functional polymorphisms and haplotype distributions of a set of common polymorphisms in the 17beta-hydroxysteroid dehydrogenase type 2 gene (HSD17B2), the aromatase cytochrome P450 gene (CYP19A1), estrogen receptor alpha (ESR1), estrogen receptor beta (ESR2), progesterone receptor (PGR) and CRC risk. Findings from the proposed study of estrogen and progesterone will enhance our understanding of these lifestyle and genetic factors in relation to colorectal carcinogenesis.
{ "pile_set_name": "NIH ExPorter" }
Granule mobilization and degranulation are important aspects of neutrophil function in acute inflammation and host-defense against microbial infection. When neutrophils are activated, their cytoplasmic granules are mobilized and transported to the plasma membrane for exocytosis and for fusion with phagosomes. There is compelling evidence that microtubules have a role in the translocation of neutrophil granules as they do in the directed movement of organelles in other types of cells. Although recent studies have revealed much about the biochemical machinery of microtubule-based organelle translocation systems, basic questions concerning the control mechanisms which govern these systems remain unanswered. These questions are particularly relevant to an understanding of neutrophil degranulation and exocytosis because these processes are highly regulated. This proposal will examine the molecular basis for regulated granule- microtubule interactions in human neutrophils. Studies will be directed at defining reversible modifications to ATPase proteins (e.g., kinesin and cytoplasmic dynein) that propel organelles along microtubules in resting and activated neutrophils. Studies will also examine post- translational modifications of tubulin that occur in response to neutrophil activation and their effects on granule-microtubule interactions. Kinesin and kinesin-associated proteins will be isolated from resting and activated human neutrophils by affinity chromatography using antibodies produced against peptides based on different regions of the amino acid sequence of Drosophila kinesin. Purified proteins will then be analyzed for neutrophil activation-induced changes in phosphorylation and electrophoretic mobility as well as changes in microtubule-activated ATPase activity and in their capacity to move microtubules in an in vitro system. Analogous studies will also be carried out in which cytoplasmic dynein and dynein-associated proteins will be isolated by immunoaffinity chromatography and examined for post- translational modifications associated with neutrophil activation. Tubulin phosphorylation, as well as tyrosination, acetylation, and glutamylation of the alpha-tubulin subunit, in the microtubules of resting and activated neutrophils will also be examined. Neutrophil activation-dependent changes in the relative proportions of tubulin isoforms will be determined by quantitative immunoblotting. Microtubules, polymerized in vitro from different isoforms of tubulin, will be examined for their capacity to enhance the ATPase activity of purified neutrophil kinesin and dynein and for their binding affinity for neutrophil granules isolated by density gradient centrifugation. It is anticipated that these studies will expand our understanding of basic mechanisms that control neutrophil degranulation and could define potential targets for the design of novel anti-inflammatory agents.
{ "pile_set_name": "NIH ExPorter" }
Bulinus truncatus rohlfsi (Ghana strain) has been successfully adapted and characterized at Southwest Foundation for Research and Education, San Antonio, Texas. At present, we are in the sixth generation of snails. Histological characterization of Schistosoma haematobium (Ghana strain), as well as normal histological studies of B. truncatus rohlfsi (Ghana strain), is being completed. These studies will determine the histopathological effect of the infection on the snail host and will determine the route and time relationship of the parasite migration in the snail host.
{ "pile_set_name": "NIH ExPorter" }
DESCRIPTION (Applicant's abstract): The fundamental goal of this proposal is to investigate cellular mechanisms associated with plasticity in the carotid body (CB) chemoreceptors. Specifically, we propose to investigate cellular mechanisms which underlie putative hypoxia-induced functional recovery of impaired CBs in rats treated with perinatal hyperoxia. In rats, perinatal hyperoxia (60 percent O2 for the first month post-partum) impairs the ventilatory response to hypoxia, an effect that persists into adulthood. This is due to impaired CB function and includes a reduction in CB size and number of chemoafferent axons from the petrosal ganglion (PG). It is postulated that perinatal hyperoxia suppresses normal maturation of the CB and PG chemoafferent neurons within a critical developmental window. Once this developmental window has ended, normal mechanisms of developmental plasticity cannot proceed. Recent studies indicate that 1 week of sustained hypoxia (SH) induces significant recovery of the hypoxic ventilatory response, and preliminary data suggest that this recovery may be CB-mediated. Our central hypothesis is that SH-induced recovery of ventilatory function arises from plasticity in the CB and its chemoafferent neurons. To test this hypothesis, adult rats subjected to perinatal hyperoxia, will be exposed to SH for up to 4 weeks. Predictable outcomes should determine if SH-induced recovery of CB function is permanent. Measurements in SH-treated and control groups will include assessment of CB function by carotid sinus nerve recording, immunocytochemical analysis and/or mRNA analysis of functional proteins (phosphorylated cyclic-AMP response element-binding protein, tyrosine hydroxylase, brain-derived neurotrophic factor, glial cell line-derived neurotrophic factor, vascular endothelial growth factor) and evidence of morphological recovery in the CB and PG. This project should provide new information on SH-induced neuroplasticity of the CB and PG in both perinatal hyperoxia treated and normal animals and may suggest methods to treat children with impaired arterial chemosensitivity due to perinatal hyperoxia.
{ "pile_set_name": "NIH ExPorter" }
This subproject is one of many research subprojects utilizing the resources provided by a Center grant funded by NIH/NCRR. The subproject and investigator (PI) may have received primary funding from another NIH source, and thus could be represented in other CRISP entries. The institution listed is for the Center, which is not necessarily the institution for the investigator. This project investigates a fascinating structure-function relationship in tagatose-1,6-biphosphate (TBP) aldolase from Streptococcus pyogenes. TBP aldolase can cleave, apart from its own substrate TBP, three other bisphosphorylated D-hexoses: fructose-1,6-bisphosphate, sorbose-1,6-bisphosphate and psicose-1,6-bisphosphate. These four sugars are diastereoisomers and differ only in stereochemistry at carbon 3 and at carbon 4 with respect to the configuration of their hydroxyl groups. It is the elucidation of this intriguing nonspecific cleavage mechanism that prompted our structural and enzymatic study of the TBP aldolase from S. pyogenes. A number of structures of various enzymatic intermediates have been solved and are being analyzed in the light of detailed kinetic studies.A second objective is to examine the role of a highly homologous orthologue of this enzyme that binds directly a regulator of virulence genes expression in S. pyogenes. The TBP aldolase orthologue is thought to be able to convey environmental cues to the gene expression regulation machinery via its binding to the regulator. The hypothesis is one of binding by the enzyme with its natural substrate dihydroxyacetone-P induces conformational changes in the enzyme's structure that allow sequestration of the regulator. Structural studies to examine this hypothesis of orthologue function are under way.
{ "pile_set_name": "NIH ExPorter" }
This subproject is one of many research subprojects utilizing the resources provided by a Center grant funded by NIH/NCRR. The subproject and investigator (PI) may have received primary funding from another NIH source, and thus could be represented in other CRISP entries. The institution listed is for the Center, which is not necessarily the institution for the investigator. The long-term goal of this research is to identify and characterize molecular mechanisms through ubiquitylation regulates tumorigenesis. The central hypothesis of the proposal is that specific E3 ligase complexes mediate the ubiquitylation of the molecular scaffold Kinase Suppressor of Ras 1 (KSR1) to control its half-life, and signaling. KSR1 is a scaffold for the Raf/MEK/ERK kinase cascade. We observed that KSR1 interacts with the components of ubiquitin E3 ligases including CUL4, DDB1, VPRBP (DCAF1), EDD1, and BRUCE (BIRC6). Overexpression of CUL4A promotes ubiquitylation of KSR1. The kinase DYRK2, which interacts with the DDB1-VPRBP complex, phosphorylates katanin making it a substrate for ubiquitylation by the ligase. Notably, the consensus sequence for substrate phosphorylation by DYRK2 is found surrounding Thr274 and Ser392 in KSR1. These data suggest that the ability of KSR1 to affect Ras-induced tumorigenesis is regulated by its ubiquitylation by specific E3 ligases. We will test this hypothesis by characterizing the interaction of KSR1 with E3 ligase components and determining whether the ubiquitylation of KSR1 affects cell transformation and tumorigenesis by oncogenic Ras. These studies should reveal novel mechanisms underlying cellular control of proliferation and transformation. Such knowledge should open new avenues for exploration and understanding of the molecular mechanisms affecting cancer susceptibility in humans.
{ "pile_set_name": "NIH ExPorter" }
3. Methodology and Statistics Core Aim 2: Provide a structure for methodological and statistical support as an integral part of the Center to assist in our proposed R01 and pilot work, as well as to develop appropriate analytic protocols for future research, including proposals to NIMH and other agencies. D.3.a. Mission. The mission of the Methods and Statistics (M&S) Core is to provide Advanced Latino Disparities Center investigators with an understanding of methodological techniques necessary to ensure rigorous research and to facilitate the professional development of researchers interested in health care disparities. This goal will be achieved by embedding the expertise of experienced qualitative and quantitative methodologists into the activities of the projects. Members of the M&S Core are integral participants in the R01s and pilots. The M&S Core will provide expertise to the Center in four areas: research design and methodology, database management, psychometrics and statistical analysis, and qualitative research methods. This core brings together the resources of its faculty to emphasize the contribution of both qualitative and quantitative methods that have been combined throughout the projects of the Advanced Latino Disparities Cente
{ "pile_set_name": "NIH ExPorter" }
This study was initiated to evaluate rigorously the clinical efficacy and specificity in migraine headache of operantly learned hand temperature control via skin temperature biofeedback. Several patients have begun such biofeedback-assisted training or noninstrumented autogenic relaxation training, while most others are still in the baseline period monitoring headache activity.
{ "pile_set_name": "NIH ExPorter" }
Atherosclerotic disease is the leading cause of death, justifying continued effort to identify the determinants of this disease. Among the priorities for the NHLBI Division of Cardiovascular Diseases is the need to better characterize inherited predispositions to atherosclerosis in the context of understanding biological pathways of inflammation that could lead to biomarkers for prediction, early detection, and prognosis. The cellular adhesion pathway is one promising new avenue of research aligned with DCVD strategic goals. Although circulating adhesion protein levels predict increased risk of incident CHD and subsequent events in patients with acute coronary syndromes, existing literature focuses only on a small subset of proteins and genes comprising the adhesion pathway and thus few studies have assessed the combined effects of adhesion components. Therefore, we hypothesize that the adhesion pathway, as modeled by individual protein levels and a composite protein score, is associated with subclinical atherosclerosis. Furthermore, we hypothesize that variants in adhesion genes account for a significant portion of the variation in adhesion protein level and that these variants are associated with subclinical atherosclerosis. To test these hypotheses, we plan to expand phenotyping and genotyping of adhesion pathway components, incorporating results from previous genetic studies, including GWAS data, in the Multi-Ethnic Study of Atherosclerosis (MESA). Our three study aims are Aim 1) To test the hypothesis that the adhesion pathway is associated with subclinical atherosclerosis, we will analyze cross-sectional associations of 15 de novo adhesion protein levels and a composite adhesion protein score with subclinical atherosclerosis, Aim 2)To test the hypothesis that variants in adhesion genes account for a significant portion of the variation in adhesion protein level and that these variants are associated with subclinical atherosclerosis, we will assess the contribution of polymorphisms within genes directly connected to the measured protein with the corresponding circulating protein levels and we will analyze the association of these polymorphisms and longitudinal change of subclinical disease, and Aim 3) To replicate the strongest findings we will analyze the association of the most promising three proteins and 384 SNPs using 3,437 MESA subjects not included in Aims 1 & 2. Cellular adhesion is critical in the pathophysiology of atherosclerosis and a better understanding of these mechanisms on subclinical disease may have important consequences for characterizing asymptomatic individuals and for preventing and improving care of patients with atherosclerosis. Therefore, our application expands the phenotyping and genotyping of adhesion pathway components in existing stored MESA study samples to understand the relationship of adhesion on the pathogenesis of atherosclerosis. Our ability to determine the relationships among protein levels, genetic variants, and subclinical disease in substantial numbers of subjects from four ethnic groups will greatly enhance our understanding of the roles these proteins play in the pathogenesis of disease.
{ "pile_set_name": "NIH ExPorter" }
The goal of this proposal is to assess the feasibility of an all-molecular method for activity-dependent feedback control of neuronal activity. We propose to generate calcium sensitive light emitting molecules (bioluminescent enzymes, luciferases) that drive light sensing optogenetic elements (channels or ion pumps, opsins) to control membrane voltage at the level of single cells for positive and negative feedback control. By adjusting calcium sensitivity and molecule location, light production can be made specific to large events such as bursts, or sensitive to individual spikes or single channel activity. By coupling these new luciferases to opsins, highly specific sensing of calcium at its source will trigger opsin activation for augmenting or suppressing neuronal activity, allowing a high degree of temporal and spatial precision in feedback control. Goals will be achieved by pursuing three aims: 1) Developing a calcium sensing split luciferase with significantly improved speed, brightness and range of sensitivity; 2) Targeting these new molecules to subcellular domains to enable highly specific biological outcomes; 3) Linking these new innovations to optogenetic readouts. Our strategy is non- invasive and it could be applied to large-scale manipulation of neural activity in behaving animals or in humans, where non-invasive, rapid feedback control of neuronal activity could be used to regulate clinically relevant activity in the brain. Our experiments are early stage, require proof of principle feasibility studies, but they have the potential to lead to a novel strategy to regulate activity only during periods of abnormal neuronal firing, such as attenuating runaway activity or amplifying local fluctuations. The molecular tools generated towards these feasibility experiments will be highly valuable in their own right, and achieving the goal of neural activity regulated self-control of neurons will be transformative.
{ "pile_set_name": "NIH ExPorter" }
The decline in adaptive immunity, T lymphocyte output and the contraction of the T cell receptor (TCR) repertoire with age is largely attributable to thymic involution. The loss of thymic function with age may be due to diminished numbers of thymic progenitors and the loss of critical cytokines and hormones from the thymic microenvironment. We have previously demonstrated that the orexigenic peptide hormone, ghrelin, and the anorexigenic hormone, leptin, are expressed by immune cells and regulate T-cell activation and inflammation. Our current work has demonstrated that both ghrelin and ghrelin receptor expression within the thymus diminishes with progressive aging. Infusion of ghrelin into old mice significantly improves the age-associated changes in thymic architecture and thymocyte counts with an increase in recent thymic emigrants (RTEs) and improvement of TCR diversity of peripheral CD4+ and CD8+ T cells. The ghrelin-induced thymopoiesis during aging was associated with enhancement of early thymocyte progenitors (ETP) and bone marrow-derived lineage negative sca-1, c-kit high (LSK) cell. Furthermore, the ghrelin and GHS-R deficient mice displayed enhanced age-associated thymic involution with reduced thymopoiesis and contraction of LSK hematopoietic stem cell compartment. The accelerated thymic involution observed in GHS-R knock out mice was reflected in major perturbations in the TCR repertoire of peripheral T lymphocyte subsets. Similarly, leptin infusion into old mice demonstrated similar effects on thymic progenitors and thymic involution as was observed with ghrelin and the pituitary hormone, growth hormone (GH); however, these anorexigen-induced changes appear to be mediated via distinct signaling pathways. Our findings demonstrate a novel role for these hormones and their receptors in thymic biology and suggest a possible therapeutic benefit of harnessing these hormonal pathways in reconstitution of thymic function in elderly and in immunocompromised subjects. Moreover, we have found that these hormones modulate a variety of leukocyte functions and support the existence of a functional immunoregulatory network involving orexigenic and anorexigenic hormones in controlling immune cellular activation and differentiation, hematopoiesis and cell survival.
{ "pile_set_name": "NIH ExPorter" }
The goal of this proposed study is to elucidate the molecular mechanisms by which hepatocytes transport secretory and endocytosed proteins to defined cytoplasmic locations. Specifically, we will test the hypothesis that the microtubule cytoskeleton and its associated ATPases, or motor enzymes, play a major role in the organization, transport, and targeting of different vesicle populations within the hepatocyte. Numerous studies have implicated microtubules in vesicular transport and liver pathology. However, previous approaches have been largely indirect and have relied on drug perturbation either in intact animals or perfused organ systems and have yielded provocative but incomplete and conflicting results. Thus, it has not been established that microtubules and associated ATPases are required for these movements. At present, the mechanisms by which the hepatocyte discriminates between secretory granules, endosomes, lysosomes and other vesicular compartments to direct them to either sinusoidal or canalicular surfaces with precision and efficiency are totally undefined. Our goal is to conduct a definitive and novel study on the role of the microtubule-based cytoskeleton in vesicular trafficking within the hepatocyte using state of the art cell biological techniques.The proposed has three specific aims. First, we will examine how microtubules are organized and polarized within the hepatocyte in respect to the sinusoidal and canalicular surfaces by: a) confocal- immunofluorescent and electron microscopic examination in conjunction with computer-aided reconstruction morphometry; b) microtubule polarity assays of cultured hepatocytes. Second, we will determine what secretory and endocytotic components move along microtubules in cultured hepatocyte couplets by combining microinjection of fluorescent probes and unique inhibitory antibodies or drugs with computer-enhanced, fluorescent, video microscopy. Third, we will test the participation of microtubules and associated motor enzymes (kinesin, dynein, dynamin) in vesicle transport through morphological and biochemical manipulation of permeabilized and homogenized hepatocytes, and cell-free systems using purified vesicular and cytoskeletal components. The technology and experiments described in this proposal are unique to the study of vesicular transport in hepatocytes. They will expand greatly our understanding of how liver cells secrete/excrete proteins and how these critical processes are disrupted by drugs or diseased states such as cholestasis and alcohol-induced cirrhosis.
{ "pile_set_name": "NIH ExPorter" }
Administrative Core Abstract The Administrative Core will coordinate the scientific and administrative activities of this HIVRAD Program, ensuring that the Projects and Cores function well together. In particular, the Core will be responsible for managing HIVRAD finances, managing scientific progress, and coordinating Projects and Cores through scientific meetings and communications, as well as ensuring full financial and regulatory compliance. A primary activity of the Core will be monitoring HIVRAD progress relative to milestones, and measuring this progress against available funding to ensure resources are used efficiently. The Core will coordinate the efforts of a Scientific Advisory Board (SAB) that will review the progress of HIVRAD projects and cores at the HIVRAD annual meeting. Core staff are part of an existing highly experienced and integrated management team, which will enable immediate start-up and rapid progress. This teamwork will accelerate advancement toward the Program's goals, thus maximizing the opportunities to positively impact HIV-1 prophylactic vaccine development. The Specific Aims of the Administrative Core are as follows. Aim 1. Provide overall management, coordination and supervision of the HIVRAD program. Aim 2. Ensure timely financial accounting and reporting. Aim 3. Ensure compliance with all institutional, state, and federal research guidelines. Aim 4. Develop and implement HIVRAD program communication, publication, and data sharing plans. Aim 5. Provide statistical support for all data analyses.
{ "pile_set_name": "NIH ExPorter" }
(a) Demonstration of a neurotrophic factor in crude extracts of the CNS for the maintenance of the acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) contents of the preganglionically denervated superior cervical ganglion (SCG) of the cat will be followed up by: (a) investigation of its mechanism of action and (b) initiation of a program to isolate and characterize the action factor. The mechanisms by which aprotinin and continual anesthesia with sodium pentobarbital oppose the fall in AChE and BuChE in denervated SCG will also be studied. The investigation of the site of origin of BuChE in the ganglion cells of the cat SCG will be continued. The electron microscopic (EM) localizations of AChE and BuChE in the carotid body, retina, olfactory tubercle, adrenal medulla, and other selected tissues will be studied. (2) New approaches will be used in attempting to localize the sodium dependent high affinity choline uptake (SDHACU) system histochemically. (3) A histochemical method will be sought for the localization of neurotoxic esterase (NTE), the enzyme whose inactivation has been proposed as the mechanism for the production of delayed neurotoxicity (DN) by certain organophosphates. (4) Histochemical investigation of the distinction between monoamine oxidase (MAO) A and B will be continued.
{ "pile_set_name": "NIH ExPorter" }
The goal of the Transgenic/knock-out Mouse Core (TMC) is to produce genetically modified mice as in vivo models for exploring the normal function of genes, and the role of genetic mutations in the etiology and treatment of cancer. The range of modifications that can be introduced into the genome of the laboratory mouse include integration of exogenous DMA (transgenes) useful in gain-of-function and cell labeling, and the precise excision (knock-out) or alteration (knock-in) of gene function that can be controlled both tissuespecifically and temporally. Transgenic and knock-in/knock-out mice are often the logical extension of studies initiated in vitro, and provide a model system with greater anatomical and physiological relevance to human disease. Genetically modified mice have been essential tools in elucidating the molecular underpinnings of many types of cancer, and supplement the traditional sub-cutaneous xenograft model for testing new anti-cancer therapies. During the most recent 12-month reporting period (January 2005 to December 2005) nine Cancer Center members from 3 programs and one non-aligned member used the transgenic/knock-out mouse shared resource. The number of users with peer-reviewed funding represented 66% of users overall. Annual budget for the core is $269,657, with 68% from the institution, 13% from user fees, and 19% ($50,000) requested from the CCSG.
{ "pile_set_name": "NIH ExPorter" }
Post-menopausal osteoporosis, a global public health problem, has for decades been attributed solely to declining estrogen levels, and although follicle stimulating hormone (FSH) levels rise sharply in parallel, a direct effect of FSH on the skeleton has never been explored. The only ascribed function of FSH is ovarian estrogen secretion. We speculate that, in addition to declining estrogen, FSH drives the decreases in bone mass during the early menopause by stimulating the osteoclast, the cell that resorbs bone. Mice devoid of FSH or its receptor do not display hyper-resorption or bone loss despite being severely hypogonadal. That FSH is pro-resorptive is supported by in vitro evidence for a G-protein coupled FSH receptor (FSHR) on the osteoclast. FSH also enhances the release of the osteoclastogenic cytokine TNFa from osteoclast precursors, and additionally, promotes osteoclast survival. The hypothesis emerging from our study, supported by the tight correlations between bone mass and serum FSH in humans, is that circulating FSH directly stimulates osteoclastic bone resorption. We will therefore investigate in Specific Aim 1 whether FSH causes bone loss in vivo independently of lowered estrogen. For this, we will administer or over-express FSH in mice lacking the two estrogen receptors, ER??-/-, as well as GnRH-deficient hpg mice. We will also examine whether selective FSHR deletion in the osteoclast will prevent ovariectomy-induced bone loss, and whether transgenic reconstitution of the FSHR in FSHR-/- osteoclasts will restore resorptive activity. In Specific Aim 2 we determine the mechanism of the FSH effect. We will first study the mechanism of FSH-induced TNFa expression, and then, using TNFa-/- mice, elucidate if the entire effect of FSH is TNFa-dependent. Finally, using Akt-deficient cells, we will also determine whether the pro-survival action of FSH is Akt-mediated. If FSH is proven to be pro-resorptive in vivo, we envisage attenuating FSH in humans to a skeletal advantage without compromising ovarian function, for example by a monoclonal antibody. The latter premise arises from our observation that FSH haploinsufficiency in mice increases bone mass, while sparing the ovaries. The significance of this work thus lies not only in our challenging an archetypal paradigm, estrogen deficiency, as being the full explanation for menopausal bone loss, but also in establishing that pituitary hormones, such as FSH, act beyond traditional target endocrine organs.
{ "pile_set_name": "NIH ExPorter" }
The purpose of this research is to determine how radiation therapy alters the various components of solid tumors that may affect the immune system's ability to mount effective anti-tumor responses. The goal of these studies is to gain a better understanding of the basic mechanisms involved in radiation induced changes in the tumor microenvironment to allow more effective treatment strategies combining radiation and immunotherapy. The differences in these two therapy modalities make it highly likely that their combined use could have synergistic effects on tumor destruction. Radiation is highly effective at killing large numbers of tumor cells and controlling primary disease, however, it is not easily used for the treatment of dissem- inated metastatic disease. In contrast immunotherapy, because of its specificity and systemic nature, has the potential to be less toxic to normal tissues and to reach distant metastases. However, radiation therapy may itself be damaging to critical cells of the immune system, particularly in multi-radiation dose protocols in which subsequent treatments may destroy tumor infiltrating lymphocytes. We have shown that radiation induced tumor cell death can provide increased tumor antigen to stimulate immunity, and radiation can alter the tumor vasculature making it more prone to host cell infiltration. These tumor infiltrating host cells produce many cytokines, which can stimulate changes in the tumor vasculature and tumor stroma and thus alter growth potential and radiation sensitivity. For example, we have shown that interleukin-12 (IL-12), produced within the local tumor microenvironment, causes marked changes in the tumor vasculature by preventing the upregulation of vascular endothelial growth factor (VEGF) receptor 3 leading to enhanced immune cell infiltration and delayed tumor growth or rejection. In this proposal we plan to determine the mechanisms responsible for these IL-12 induced changes in tumor vasculature and in the immune cell infiltration and function, and determine the role of interferon-gamma (IFN-g) in these processes. Due to the importance of IFN-g in these IL-12 induced changes and in alterations that render tumor cells more radioresistant, we will also determine the mechanisms by which IFN-g signally mediates radioresistance and how these affect immune sensitivity of tumors. The information we obtain will be useful in improving the combination of radiotherapy and immunotherapy to more effectively treat patients with cancer. [unreadable] [unreadable] [unreadable]
{ "pile_set_name": "NIH ExPorter" }
Prematurity, enteral formula feeding, and bacterial colonization are three major risk factors for neonatal necrotizing enterocolitis (NEC). The hallmark pathologic findings in NEC are intestinal mucosal injuries. However, the etiology and pathogenesis of NEC remain unclear. Acetic acid, propionic acid and butyric acid are short chain fatty acids (SCFAs) which are produced mainly in the colon by bacterial fermentation of undigested carbohydrates. In physiological condition, the production of SCFAs in the bowel is very important for energy salvage and crucial for gastrointestinal adaptation and maturation. However, excessive concentrations of SCFAs (especially butyrate) can deregulate the normal process of apoptosis and may cause disruption of the intestinal barrier. Once the intestinal barrier is disrupted, the inflammatory cascade may be activated, which can induce further injury to the intestinal mucosa. Therefore overproduction/accumulation of SCFAs in the intestinal lumen may play a pivotal role in the pathogenesis of NEC. Our previous study and our preliminary data suggest that butyrate in physiological concentration promotes the intestinal barrier function by regulating the assembly of tight junctions via activating AMP-activated protein kinase (AMPK). Excessive luminal SCFAs, especially butyrate, however may induce severe apoptosis and disrupting mucosal barrier. The overall objectives of this proposal are to elucidate the molecular mechanisms underlying the effects of butyrate on intestinal barrier function. Firstly, we will determine if AMPK activation is the primary mechanism mediating the effect of butyrate at physiological concentration on the assembly of tight junction proteins by using a well established Caco-2 cell monolayer model. Then, by using the same Caco-2 cell monolayer model, we will determine if the augmented apoptosis induced by butyrate explains the detrimental effect of excessive butyrate on the intestinal barrier. PUBLIC HEALTH RELEVANCE: Short chain fatty acids (SCFAs) in physiological concentration promote the intestinal barrier function. However, excessive luminal SCFAs, especially butyrate, can cause intestinal injury by inducing severe apoptosis and disrupting mucosal barrier function. The project of this proposal is to further elucidate the molecular mechanisms underlying the effects of butyrate on intestinal barrier function, which may help us understand the role of SCFAs in the pathogenesis of neonatal necrotizing enterocolitis (NEC).
{ "pile_set_name": "NIH ExPorter" }
The Administration & Outreach Core supports SPORE projects and investigators by managing SPORE resources, quality control, and communication and outreach, including fostering interaction among SPORE components and collaborators, other SPORES, the patient and advocate community, and the NCI. This management and support is accomplished through a series of oversight committees and organized administrative and scientific meetings of SPORE investigators, institutional representatives and external advisors. Specific functions of the Core include: ? To administratively manage and coordinate all SPORE-related research ? To monitor and manage financial resources ? To create and prepare documents and reports to ensure compliance with federal regulations and reporting requirements ? To administer the Developmental Research (pilot project) and Career Development Programs ? To organize all meetings, seminars, and travel related to Breast Cancer SPORE activities ? To serve as a point of contact to all VICC fundraising activities focused in breast cancer ? To support and coordinate all internal and external collaborations ? To serve as a main point of contact to biotechnology and pharmaceutical companies Carlos L. Arteaga, MD, and Jane Kennedy, MSSW, will serve as co-Directors of the Core. Arteaga reports directly to Jennifer Pietenpol, PhD, Director of VICC, and has input and assistance from the Deputy Director (Daniel Beauchamp, MD) and the Associate Directors for Basic Science (Scott Hiebert, PhD) and Cancer Epidemiology, Prevention & Control (William Blot, PhD). Arteaga provides advice to Dr. Pietenpol on programmatic directions and allocation of VICC resources for research in breast cancer. Since 2006, Ms. Jane Kennedy has served as Manager of Patient Advocacy in the VICC Office of Patient & Community Education (OPACE). Ms. Kennedy's role includes recruiting, training and coordinating the survivors and caregivers who contribute to and support the Research Advocacy Program of VICC, including the Breast SPORE.
{ "pile_set_name": "NIH ExPorter" }
DESCRIPTION: Alzheimer?s disease (AD) is the most costly and devastating neurodegenerative disorder of the 21st century. To conquer AD, therapies must be developed to prevent the neuropathological manifestations of the disease. It is widely held that accumulation of beta amyloid (AB) plaques in AD brain causes oxidative damage and inflammatory responses involving glial cell activation that, in turn, leads to neuronal cell death. Many factors, including apolipoproteins, phospholipids and cholesterol, can influence aggregation of AB and its cytotoxic actions. There is evidence for the roles of plasma membrane lipids and G protein-coupled receptor functions in the cytotoxic effects of AB in the AD brain. The major loci of this Program Project Grant (PPG) are 1) to examine the effects of AB and oxidant stressors on neural cell functions relevant to the AD phenotype, including activation of phospholipases, cyclo-oxygenases and G protein-coupled receptors, and 2) to determine relationships between cholesterol homeostasis, lipoproteins and AD. This highly integrated PPG, directed by Dr. Grace Sun, consists of an Administrative Core, a Cell Culture Core and three research projects. Project #1 (G. Sun, PL) will determine the effects of AB and lipoproteins on oxidative and inflammatory responses in neural cells mediated by phospholipases A2. Project #2 (G. Weisman, PL) will determine the effects of AB and lipoproteins on G protein-coupled P2Y2 nucleotide receptor functions in glial and neuronal cells. Project #3 (G. Wood, PL) will determine how AB affects cholesterol homeostasis, transport and distribution in cell membranes and subcellular organelles. By understanding the role of lipids, lipoproteins, and receptor-mediated cell signaling pathways in AD pathology, the proposed studies will provide important new information that will impact the development of effective pharmacotherapies for this debilitating disease.
{ "pile_set_name": "NIH ExPorter" }
Numerous clinical observations indicate that in certain patients alloantibodies probably contribute to rejection. Although some investigators have attributed a causal relationship to this association, there are still insufficient data to evaluate critically the mechanisms of such antibody-mediated acute allograft injury. Presently, the availability of inbred "knock out" mice, monoclonal antibodies to critical mediators, and soluble recombinant receptors provide the means to establish a causative role of antibody in the in vivo pathogenesis of acute graft rejection. Recent in vitro studies have demonstrated that antibodies and complement can initiate dynamic interactions among endothelial cells, macrophages and platelets that are mediated by adhesion molecules and their ligands. The potential role of these receptors and their ligands has not been examined in acute alloantibody-mediated rejection. The hypothesis to be tested in this proposal is that alloantibody and complement mediate acute cardiac rejection by activating endothelial cells, macrophages and platelets through ,expression of P-selectin and von Willebrand factor. The proposed studies are based upon our extensive preliminary data from cardiac allograft models in complement deficient rats as well as immunoglobulin knock out (IgKO), C4, C3 and CR2 KO mice. The specific aims of this project will test our hypothesis by demonstrating that: 1. Passive transfer of complement-activating classes and subclasses of mAbs specific for donor MHC antigens can reconstitute cardiac allograft rejection in IgKO mice and cause the upregulation of P- selectin, von Willebrand factor, and MCP-l in endothelial cells. 2. Passive transfer of non-complement-fixing subclasses of mAbs specific for donor MHC antigens will modulate in a dose-dependent manner cardiac allograft rejection induced in IgKO mice by complement-activating subclasses of mAbs with the same specificity. 3. Activation of endothelial cells, B-cells, platelets and macrophages by antibody mediated acute rejection is inhibited in C4, C3 and CR2 KO mice resulting in prolonged cardiac allograft survival. 4. P-selectin augments activation of endothelial cells, platelets and macrophages in allograft rejection.
{ "pile_set_name": "NIH ExPorter" }
Abstract Therapeutic management of metastatic castration-resistant prostate cancer (mCRPC) remains a major clinical challenge. Localized or organ-confined prostate cancer can be cured by surgery, or a combination of androgen deprivation therapy (ADT), radiotherapy and/or chemotherapy. However, tumor recurrence occurs in some men due to the development of ADT-resistant clones which greatly contributes to the lethality of prostate cancer. Aggressive mCRPC frequently metastasizes to distant organs such as bone. As a result treatment becomes challenging and survival rate drops. Hence, there is an urgent need to develop new therapies to cure advanced prostate cancer. Development of aggressive disease is favored by activation of several `escape pathways' among which AR- coactivators such as steroid receptor coactivator-2 (SRC-2/NCOA2) plays a critical role promoting the survival and rapid metastasis of CRPC. SRC-2 promotes a ?metabolic switch? in advanced tumors that predispose them to be dependent on `glutamine' to generate energy and macromolecules required for survival and metastatic growth. Ablation of SRC-2 suppresses prostate tumor survival and metastasis in vivo, indicating this may be a viable approach to treat advanced patients. So our objectives in this proposal are (1) to investigate the mechanisms that promote increased glutamine metabolism in tumors, (2) identify the upstream signaling events that stimulate SRC-2 transcriptional responses to regulate this metabolic reprogramming, and (3) examine the therapeutic benefits of targeting this `metabolic switch' to block cancer progression and metastasis. During the K22 award I expect to identify the molecular links between cellular metabolism and oncogenic events in mCRPC, and examine the potential benefits of targeting this pathway to selectively impair prostate cancer metastasis. The study will make novel insights depicting the altered metabolic pathways and underlying mechanisms promoting the emergence of CRPC, which may lead to the discovery of potential prognostic tools for early detection of clinically significant disease.
{ "pile_set_name": "NIH ExPorter" }
PROJECT SUMMARY/ABSTRACT Mouse primary visual cortex (V1) is capable of encoding visual stimuli and undergoing bidirectional plasticity coincident with patterned visual experience. The prefrontal anterior cingulate cortex (ACC) receives input from visual cortex, but mechanisms of visual encoding and experience-dependent plasticity in ACC are not well- understood. The goal of the proposed research is to define how ACC encodes and adapts to patterned visual experience. During the K99 phase, I will pair in vivo and ex vivo recordings with optogenetic and chemogenetic circuit manipulations to study how ACC microcircuits encode visual input. K99 training in two-photon calcium imaging will enable longitudinal studies of how individual neurons adapt to visual experience during the R00 phase. As abnormal sensory processing is a common feature of autism spectrum disorder, further R00 research will use approaches developed during K99 training to assess ACC microcircuit function in single-gene mouse models of autism. My long-term career goals are to advance basic knowledge of visual processing and to improve the lives of individuals with neurodevelopmental disorders through a collaborative program of basic and translational research. To achieve my goal of securing a tenure track faculty position as an independent researcher in neuroscience research, I have developed a comprehensive career development plan with the support of co-mentors Dr. Ben Philpot and Dr. Spencer Smith (UNC) and collaborators Dr. Paul Manis (UNC) and Dr. Jeff Gavornik (Boston University). This plan includes a) technical training in two-photon imaging, b) collaborative projects, c) training in presentation skills, d) training in mentoring skills, e) training in grant writing, f) career guidance, g) clinical exposure, h) formal coursework, i) teaching opportunties, and j) progress monitoring and evalulation.
{ "pile_set_name": "NIH ExPorter" }
As a health communication, multimedia and graphic design firm, we, like many others, are consumers of high quality royalty-free stock photography for health education materials. Unfortunately, there is a dearth of readily-available images depicting health conditions and behaviors that match characteristics of target audiences defined by race, socio-economic status, age, weight, medical conditions, and physical disabilities. This shortcoming is a theoretic and practical problem for health education, as several common theories guiding successful health interventions predict that communication is more effective when sources and models are attractive and similar to target audiences, producing identification and perceived relevance. In Phase I, our team demonstrated the feasibility of developing a collection of photographs depicting diverse settings, health behaviors, health conditions and demographics, called Real Health, which can be used by health and media professionals to increase effectiveness of health communication by incorporating similar image referents. In this Phase II project, our team proposes to fully-develop the Real Health enterprise, an online image library for use in health communication. First, we will produce Real Health and populate it with photographs taken in Phase I and additional photographs of 240 individuals (African American, Hispanic, Native American, Asian, non-Hispanic white, and GLBT; children/teens, adults, and seniors; overweight and with mobility challenges) taken in Phase II. Potential end users (photographers and public health practitioners) will evaluate the Real Health e-commerce site and business model for efficiency, utility, user participation, and satisfaction. The Real Health website will be subjected to beta and usability testing prior to launch. Next, we will evaluate the ability of photos from Real Health to improve health education message effectiveness in a randomized pretest-posttest 3-group design. Three experimental conditions will be created, i.e., a health education message with 1) all Real Health photos that match the target audience, 2) half Real Health photos, and 3) all mismatched photos (obtained from existing commercial stock photo libraries). The health education message will advocate increased physical activity and improved diet to reduce chronic disease. Photos will be selected based on careful pretesting with target audiences. A sample of individuals who are African Americans, Hispanics, Native Americans, overweight/obese, 65 years of age or older, and with mobility challenges (n=510 per subgroup; n=3060 total) will be recruited from an Internet panel maintained by Knowledge Networks, Inc. Participants will be pretested, assigned to read a health education message in one of the conditions, and post-tested. Analyses will evaluate the effect of Real Health photos on primary (intention) and secondary (positive outcome and self-efficacy expectations) outcomes, test potential effect moderators, and explore theoretical mediators of perceived relevance, appropriateness, similarity and identification in the health education messages. KB is uniquely qualified to create and commercialize the Real Health stock photography enterprise.
{ "pile_set_name": "NIH ExPorter" }
A Severe Acute Respiratory Syndrome (SARS) pandemic, caused by re-emergence of the causative coronavirus (CoV) from an animal reservoir, is a serious threat. Induction of protective antibody by vaccination has proven the most effective intervention for highly contagious viral pathogens. The capsid proteins of viruses typically self-assemble into empty capsids known as virus-like particles (VLPs). VLPs resemble native virions immunologically. As vaccine candidates, VLPs have the advantage of being non-replicating and non-pathogenic, and of presenting host protective epitopes in a particulate and multimeric form. We have generated SARS-CoV VLPs by expression of the spike (S), membrane (M) and small envelope (E) proteins in the baculovirus/insect cell system and shown that VLP are immunogenic in rabbits. We now propose: (1) to develop efficient methods for production of highly purified SARS-CoV M+E and M+E+S VLPs from insect cell cultures, (2) to characterize the immunogenicity of SARS-CoV VLP vaccines in BALB/c mice; and (3) to determine the potency of a VLP vaccine to protect BALB/c mice and ferrets challenged with live SARS-CoV. Recent studies have shown that amino acids changes in S can mediate increased resistance to neutralization and enhancement of S pseudotype virus entry. The conserved C terminus of coronaviruses contains epitopes that contribute to enhancement. Thus, our final specific aim (4) is to determine the cross neutralizing/enhancing capacity of antisera raised in mice against VLPs of SARS-CoV strains and the human coronaviruses, OC43 and 229E. The successful implementation of the proposed studies would provide a stepping-stone toward future clinical trials with a VLP-based SARS-CoV vaccine. [unreadable] [unreadable]
{ "pile_set_name": "NIH ExPorter" }
Suicide is a complex phenomenon comprised of biological and psychological risk factors, in addition to social, environmental, and economic contributors. The most consistent of these risk factors is the presence of a psychiatric illness. One of the challenges inherent in developing strategies for suicide prevention is that suicide is a rare and unpredictable event. It is extremel important, therefore, to identify biological markers that are associated with high risk for suicide Family, twin and adoption studies strongly suggest genetic contributions to suicide; furthermore, the genetic predilection for suicide appears to be largely independent of genetic liability to mental illness. To date, many conventional association studies have been performed but have not revealed any genetic variants that could be used as reliable predictors of suicide risk. In thi application we propose using alternate approaches which we expect to be more successful in identifying such predictors. Our first approach, (which will be explored in Specific Aim 1) is related to a rarely studied mechanism of homeostatic plasticity-RNA editing of serotonin 2C receptor (5-HT2CR). Editing can generate many different 5-HT2CR isoforms which vary in their functional activity, thus enabling the 5-HT2CR-expressing neurons to respond to both environmental and genetic perturbations. Our studies, as well as studies in other laboratories, have demonstrated that 5-HT2CR editing is altered in the prefrontal cortex (PFC) of suicide victims regardless of their underlying psychiatric illness. Thus, dysregulation of editing constitutes a biological factor that is strongly associated with completed suicide. 5-HT2CR is expressed at levels that can be reliably assayed only in the brain and spinal cord. Therefore, editing cannot be noninvasively measured in living individuals in the areas relevant to suicide (i.e., PFC). We hypothesize the existence of single nucleotide polymorphisms (SNPs) that are associated with 5-HT2CR editing, and in our preliminary studies we have already identified several such candidate SNPs. Here we aim to confirm these findings and to identify novel editing-associated SNPs in a significantly larger cohort of postmortem specimens (N=583). The editing-associated SNPs can be used as a proxy for measuring editing in the brain, and therefore, as predictors for suicide risk. Our second approach (Specific Aim 2) will be to uncover SNPs that are associated with behavioral traits that constitute susceptibility factors for suicide and are, therefore, considered to represent suicide-related endophenotypes. This strategy allows the deconstruction of suicide into parts that are less etiologically and geneticall heterogeneous. To this end, we will perform genome-wide association studies (GWASs) in a large cohort (N=1,200) of demographically and genetically homogeneous young male conscripts in the Greek Army. For these individuals the results of self-reporting measures and behavioral/psychophysiological assessments of suicide-related phenotypes, as well as genomic DNA specimens, have already been obtained and are available for us. Not all genetic components that are associated with editing or with suicide-related endophenotypes are specifically related to suicide. Therefore, in our last Aim we will determine which of the SNPs identified in Aims 1 and 2 are more likely to represent suicide-specific risk factors. To achieve this goal, we will use (1) large publicly available data sets of GWASs in psychiatric patients with and without a history of attempted suicide and (2) postmortem PFC specimens from psychiatric patients who died of suicide or by other means. When confirmed, the risk alleles could be measured in peripheral blood cells, thus providing biological correlates associated with liability to suicide. Mapping these alleles will also uncover pathways that are altered in the brains of people who are prone to suicidal behavior, hence opening new venues to the understanding of pathophysiology of suicide.
{ "pile_set_name": "NIH ExPorter" }
The long-range objectives of Project 2 are to develop HlV-1 vaccine regimens that stimulate broadly directed immune responses and that block HIV-1 infection in humans and control viral replication In individuals who become infected. Although the precise correlates of immunity remain undefined, significant progress has been made in this area, and it appears that both humoral and cellular immune responses may be required for an optimal HIV-1 vaccine. In this project, we will test the hypothesis that mosaic antigens will induce increased breadth of cellular and/or humoral immune responses compared with natural sequence antigens in humans. We will also test the hypothesis that boosting responses primed with our optimal vector regimen with our stable clade 0 gp140 trimer will markedly augment antibody responses. To evaluate these hypotheses, we propose the following two Specific Aims: Specific Aim 1: To determine the optimal vector regimen in humans. Study IA: A phase 1 trial to compare natural and mosaic antigens in the context of Ad26/MVA vectors. Study IB: A phase 1 trial to compare the homologous Ad26 vector regimen with a heterologous Ad26/MVA vector regimen. Specific Aim 2: To determine the impact of a protein boost with our novel Env gpl40 trimer in humans. Study 2A: A phase 1 dose escalation trial of our novel trimeric Env gp140 protein. Study 2B: A phase 1 study to compare the best vector regimen with and without a protein trimer boost.
{ "pile_set_name": "NIH ExPorter" }
Cargo Sorting and Intralumenal Vesicle Budding by the ESCRT Complexes Membrane budding and fission is a fundamental process of eukaryotic cell biology. Endocytosis, the formation of intracellular transport and secretory vesicles, and mitochondrial fission are examples of inward budding. In the classical example of clathrin-mediated endocytosis, the cytosolic protein dynamin forms arrays on the outside of the membrane neck, and membrane fission is driven thermodynamically by the hydrolysis of GTP. The formation of multivesicular bodies (MVBs) is the prototypical example of outward budding. MVBs are formed during the maturation of endosomes destined to fuse with lysosomes, and mediate the sorting of ubiquitinated membrane proteins to the lysosome. Portions of the limiting membrane of the endosome are internalized to form intralumenal vesicles (ILVs). When the MVB fuses with the lysosome, ILV contents are degraded by lysosomal hydrolases. When ILVs are released through fusion with the plasma membrane, they are referred to as exosomes. The budding of enveloped viruses from the plasma membrane and cell division (cytokinesis) are other examples of outward budding events. Outward budding events in MVB formation, viral budding, and cytokinesis are directed from the cytosol. Since cytosol is in contact with the inside, not the outside of the neck of the nascent bud, the mechanics of membrane fission differ fundamentally from inward budding, and utilize a completely distinct protein machinery. A major breakthrough in understanding outward budding came from the identification in yeast of the ESCRT machinery responsible for MVB formation. The ESCRT machinery is conserved throughout eukaryotes, and many enveloped viruses of mammals use the ESCRT pathway to bud, including HIV-1. The closure of the membrane neck in cytokinesis also uses the ESCRT pathway. The assembly of ESCRT complexes on endosomes is triggered by the presence of phosphatidylinositol 3-phosphate (PI(3)P) and ubiquitinated cargo proteins. ESCRT-I and II directly bind to cargo, and in turn recruit ESCRT-III. There are four ESCRT-III subunits in yeast, Vps2, Vps20, Vps24, and Snf7, together with two associated ESCRT-III-like proteins, Did2 and Vps60. ESCRT-III subunits exist in the cytosol as monomers, and assemble with each other on membranes in large multimeric arrays. ESCRT-II is a Y-shaped complex that contains two copies of the Vps25 subunit, which recruits ESCRT-III by directly binding to Vps20. Vps20 binds to Snf7, comprising a subcomplex of ESCRT-III. Snf7, in turn, directly binds to the Bro1 domain of the ESCRT-associated protein Alix (known as Bro1 in yeast). The Vps20:Snf7 complex recruits the Vps2:Vps24 subcomplex to form the complete ESCRT-III complex. A subset of ESCRT-III proteins directly bind to the N-terminal MIT domain of the AAA ATPase Vps4. Vps4 is a central player in the MVB pathway that is required for the disassembly of the ESCRT-III complex. ESCRT function can be conceptually separated into two phases: cargo recruitment and concentration, followed by membrane invagination and budding. The long term objectives of this project are to: 1) determine the structures of ESCRT complexes by x-ray crystallography, abetted where necessary by electron microscopy, hydrodynamics, molecular simulations, and small angle x-ray scattering; 2) to determine how ESCRTs assemble on membranes containing PI(3)P and cargo using binding and spectroscopic techniques; and 3) to study the mechanism of ILV formation by a microscopic, spectroscopic, and structure/function approaches. ESCRT-I is a heterotetramer of Vps23, Vps28, Vps37, and Mvb12. The crystal structures of the core complex and the UEV and Vps28 C-terminal (CTD) domains have been determined, but internal flexibility has prevented crystallization of intact ESCRT-I.In FY2011, we determined a low resolution structure of ESCRT-I in solution. In FY2012 we built on this study by determining the solution structure of supercomplex formed by ESCRT-I and -II together. This is the key assembly responsible for stabilizing the neck of the membrane bud. The structural ensemble was cross-validated against single molecule Frster resonance energy transfer (FRET) spectroscopy, which suggested the presence of a continuum of open states. This study led to the most detailed model for ESCRT-mediated budding and scission to date. The physical mechanism of membrane budding by ESCRTs has been unresolved and a subject of controversy. Spurred by theoretical modeling, we tested whether ESCRTs could induce lateral lipid phase separation. Theory predicts that line tension arising between different lipid domains could promote budding. Using a supported bilayer model, we found that human ESCRT-II could assemble on supported bilayers into small clusters that induced a liquid ordered lipid domain that was not pre-exising, even in lipid mixtures that are far from the phase boundary for spontaneous segregation. The clusters bind stoichiometrically to ubiquitin and to the ESCRT-III protein VPS20. This finding helps connect theoretical analysis of ESCRTs to their biology and so clarifies the budding mechanism. Human ESCRT-I functions at multiple loci in the cell, including both endosomes and the plasma membrane. Human ESCRT-I is not a single entity, but rather a heterotetramer containing one copy each of TSG101, VPS28, VPS37 isoform A-D, and either MVB12 A-B or UBAP1. We found that the MABP domain of MVB12A and B binds to acidic lipids found at both the endosome and plasma membrane, and solved its crystal structure. This helps explain how the MVB12-containing forms of ESCRT-I can function both in endosomal cargo sorting and in plasma membrane processes such as cytokinetic abscission, HIV-1 budding, and exosome formation and release.
{ "pile_set_name": "NIH ExPorter" }
HIV associated cardiac pathology is being recognized increasingly as patients with chronic HIV infection who survive to live productive lives. HIV associated cardiomyopathy is among the most common cardiac specific manifestation of chronic HIV infection and was observed in between 6-12% of patients that succumb to AIDS. However, cardiovascular involvement has emerged as more than a pathologic curiosity, but rather as a significant cause of morbidity as individuals live longer, more productive lives with HIV. Increased use of HAART therapy has served to unmask HIV associated predispositions to cardiac involvement. Despite the increased recognition, the risks factors for, the pathogenesis of, and the specific treatments required in HIV associated cardiac disease remain unknown. The nonhuman primate model of SIV infection in rhesus macaques affords an unparalleled opportunity to study these critical features. Prior work from our laboratory has characterized the time course, the role of CIM counts, and the pleomoprhic cardiac manifestations in simian AIDS. The work has demonstrated the need to consider both viral and host factors in identifying those at increased risk and to create a consistent, reproducible model of cardiac involvement for further investigation. We have determined that macrophage-tropic strains of SIV are most commonly associated with lymphocytic myocarditis. SIV is localized to the myocardium in approximately one third of cases of cardiac involvement, and when present, always co-localizes to cells of the macrophage lineage, either tissue macrophages or cardiac dendritic cells. TNFalpha, produced by activated macrophages, mediates both up-regulation of NOS2 in cardiac myocytes and the expression of Fas (CD95) receptors leading to cardiac myocyte apoptosis. As such, we have shown that cytokines play a central mechanistic role in both reversible LV dysfunction (increased NF-kappaB NOS2 expression) and irreversible myocardial injury (Fas-FasL mediated apoptosis). In addition, lymphocytic infiltrates are frequently perivascular and associated with coronary vascular lesions characterized by endothelial activation, smooth muscle proliferation, and thrombotic occlusions, leading to acute ischemic injury. These pathological features are mirrored in the lung where lymphocytic interstitial pneumonia and pulmonary vasculopathy are observed and contribute to increased right ventricular dysfunction. We plan to explore both host and viral factors that lead to SIV transmission into the myocardium (SIV cardiotropism) and SIV mediated injury (cardiovirulence). The identified mechanisms will serve as a prerequisite for exploring specific strategies to prevent cardiac involvement in chronic SIV infection.
{ "pile_set_name": "NIH ExPorter" }
PROJECT 2 SUMMARY ABSTRACT Multiple myeloma (MM) is a slowly proliferating tumor of post-germinal center plasma cells that causes bone destruction, marrow failure, immuneparesis and renal failure and results in over ten thousand deaths each year in the USA. For many years a class of drugs termed immunomodulatory drugs (IMiDs) have been used in the clinic with remarkable activity against MM and a few other hematopoietic neoplasms (Chronic Lymphocytic Leukemia, NonHodgkin Lymphoma and Myelodysplastic Syndrome with 5q- deletion). Although many patients have a favorable initial IMiD response, most will eventually relapse with loss of sensitivity to the IMiD (acquired resistance to IMiDs). Patients also differ in their initial responses to IMiD therapy. While some enjoy `exceptional' responses to IMiD treatment (defined as time to progression of 72 months or longer) others exhibit innate resistance and thus never respond. The exact mechanism of IMiD action and resistance in MM remains elusive, although it is clear that IMiDs trigger the CRBN-dependent degradation of the hematopoietic transcription factor Ikaros. Almost every MM tumor is characterized by super enhancer-mediated oncogene dysregulation via insertion of such enhancers into the MYC locus or via recurrent IgH enhancer translocations to a number of oncogenes. Our preliminary data further indicate that many these critical super-enhancers driving oncogene expression in MM exhibit high Ikaros occupancy, and that their enhancer function is consequently often Ikaros-dependent. Our hypothesis is that tumor cell-autonomous IMiD resistance is caused by super-enhancers driving oncogene expression that retain their function in the presence of IMiDs. The work presented here studies IMiD resistance using three different approaches. First, with the goal of identifying genetic determinants of innate and acquired IMiD resistance, we will examine structural variations (primarily DNA translocations) that mobilize super-enhancers to oncogenes in patient samples that have been banked in the Mayo Clinic biobank. Secondly, we have >80 genetically annotated MM cell lines with a range of responses to IMiDs that will be tested for their response to IMiDs in terms of proliferation and changes in gene expression/protein levels (including MYC). Additionally we will examine synergies between IMiDs and a number of novel rational drug partners including next generation BET inhibitors (BETi), CBP/p300 inhibitor and a combination BETi/p300 inhibitor. Thirdly, IMiD response mediated by the inter-play between tumor cell and host will be studied in an orthotopic, immunocompetent humanized mouse model of MM in which both the tumor and the host are capable of responding to IMiDs. Our ultimate goal is to rationally optimize the activity of IMiDs, which would have a profound impact on the clinical history of MM, both avoiding IMiD resistance in newly diagnosed patients and reversing IMiD resistance in relapsed patients.
{ "pile_set_name": "NIH ExPorter" }
There were three recognized influenza pandemics in the 20th century, and historical experience suggests that another influenza pandemic is all but inevitable. Although 'colds' and seasonal influenza are clinically very different diseases from pandemic influenza, they share common transmission pathways and the community-level interventions needed to reduce both seasonal flu, common viral URIs and pandemic influenza are likely to be similar. In addition, the circulating influenza viruses causing seasonal flu are physically identical to pandemic influenza viruses, and therefore should be a good model for testing interventions before a pandemic emerges. Little research has been conducted to identify influenza prevention strategies at the household level. Hence, specific aims of this project are to compare the impact of two household-level interventions (an alcohol-based hand sanitizer with or without face masks) on six outcomes: incidence and types or strains of virologically confirmed influenza occurring in study households; rates of symptoms of influenza and viral URIs; basic reproduction number (R0), i.e. number of secondary cases generated by a single infected person in a fully susceptible household; antibiotic use practices for symptoms of influenza and other viral URIs; household member knowledge of prevention and treatment strategies for pandemic influenza and viral URIs; and rates of influenza vaccination among household members. 450 households in northern Manhattan (primarily recently immigrated Hispanics) will be randomized to three groups: control (receiving only a pamphlet on influenza prevention), alcohol hand sanitizer, and sanitizer plus face masks. Symptoms of influenza will be monitored daily for 15 months using ecological momentary assessment technology. Virologic cultures will be obtained from persons with flu symptoms (fever >100 degrees F., sore throat and/or cough). Antibiotic use practices, knowledge, and vaccination rates will be assessed by survey using pilot-tested, psychometrically sound instruments. For this cluster randomization design with randomized intervention on the household level, outcomes will be measured at the individual and household level using generalized linear mixed model for counts response with a Poisson distribution and other appropriate multivariate techniques to control for confounding. [unreadable] [unreadable] [unreadable] [unreadable]
{ "pile_set_name": "NIH ExPorter" }
Loss of voluntary control over bladder and bowel function as a result of spinal cord injury (SCI) has profound impact on the mental and physical health status and quality of life of individuals. It is estimated that 270,000 people in the USA have SCI (https://www.nscisc.uab.edu/PublicDocuments/fact_figures_docs/.pdf). Urinary retention as a result of SCI is irreversible and can be life threatening. The only available pharmacotherapy for urinary retention consists of cholinergic agonists, which have minimal efficacy and severe side effects. Consequently, many people with SCI catheterize multiple times daily to empty their bladder. Bladder catheter use is associated with increased incidence of health problems, predominately repeated urinary tract infections, sepsis, isolation, depression and hospitalization. An on demand, safe and effective, pharmaceutical alternative to catheterization would be a life-changing improvement in the daily routine of bladder management for SCI individuals, and a significant reduction in individual and community health care costs. Dignify Therapeutics is a drug development company focused on advancing novel drug therapies for patients with SCI, multiple sclerosis and similar diseases that result in voidin dysfunction. By combining novel pharmaceuticals with novel drug delivery technology, Dignify Therapeutics hopes to redefine treatment of voiding disorders and restore the dignity of voluntary excretory function in a way that mimics normal micturition. Dignify Therapeutics' lead compound, DTI-100, is being developed as the first on-demand pharmaceutical to induce bladder voiding without catheterization. Phase II studies are proposed in a translational rat model of SCI to establish the efficacy of DTI-100. Chronic SCI often results in detrusor- sphincter dyssynergia (DSD) in which the external urethral sphincter fails to properly relax during micturition, resulting in impaired voiding efficiency. Proposed studies will examine the effects of DTI-100 on bladder function in a rat SCI model with and without urethral smooth and/or striated muscle activity. These studies are essential to confirm the utility of DTI-100 in treating chronic SCI-induced voiding dysfunction in humans. To enable submission of an Investigational New Drug (IND) application and subsequent use of DTI-100 in humans, FDA-regulated drug safety and toxicology studies are required. Phase II studies are proposed to establish a toxicity and safety profile for DTI-100 that is acceptable for IND submission. Studies including repeated dose toxicology, cardiovascular, and pulmonary safety studies in two animal species will be performed to identify an acceptable dose range for human administration. Manufacture of DTI-100 drug product for use in clinical studies will also be completed. Successful completion of these studies will enable a clinical study of the safety, tolerability and pharmacodynamics of DTI-100 in humans and suggest therapeutic approaches to reduce urethral resistance in patients with high resistance (e.g. DSD).
{ "pile_set_name": "NIH ExPorter" }
The overall goal of the proposed experiments is to determine the molecular mechanisms involved in the regulation of cardiac muscle contraction by troponin and to determine its role in the genesis of familial hypertrophic cardiomyopathy (FHC). There are two major projects: In I., we will determine the fundamental role of cardiac troponin T (CTnT) in the regulation of cardiac muscle contraction. Traditionally, it has been thought that the primary role of TnT is to interact with and anchor the complex of Tnl and TnC to the actin- containing thin filaments through TnT's interactions with tropomyosin (Tm) and Tnl. Recent results from our lab, with skeletal muscle, additionally suggest that: 1) Ca/2+ binding to STnC causes an interaction between STnC and the C-terminus of STnT and causes an activation of the ATPase activity; and 2) the maximum level of ATPase activation is determined by the particular N-terminal TnT variant which is present. Due to the similarities in primary structure between CTnT and STnT, we hypothesize that CTnT may play a comparable or related role in cardiac muscle. Biochemical, molecular and physiological approaches will be used to test this hypothesis and to determine the fundamental role of HCTnT in the regulation of cardiac muscle contraction. Project II. FHC is an autosomal dominant disease which has been shown to be associated with mutations in HCTnT, and HCTnl. In this section we will attempt to learn how these mutations affect the biochemical and contractile properties of cardiac muscle and give rise to FHC. Our current working hypothesis is that mutations of CTnT and CTnI lead to changes in the interactions between the CTn subunits, or changes in their interactions with the other thin filament proteins, which in turn lead to changes in contractility and/or the Ca/2+ affinity of CTnC (which would change the intracellular [Ca/2+] transient), and that these changes could trigger the cellular mechanisms responsible for the hypertrophic process. To test this hypothesis, transgenic animal models, skinned fiber, myofibrillar, and actomyosin systems, combined with biochemical, molecular, biophysical and physiological techniques, will be utilized. The combined results from these two projects will yield important new information on the role of CTnT and CTnI in the regulation of cardiac contraction and in FHC.
{ "pile_set_name": "NIH ExPorter" }
Plague is caused by infection with the bacterium Yersinia pestis, a category A bioterrorism agent. Any strategy to protect against Y. pestis must recognize that the pneumonic form of the disease develops within days, and thus anti-Y. pestis protection has to be functional within days following an attack. This proposal presents a strategy for both rapid and long lasting protection against Y. pestis based on our experience in using adenovirus (Ad) gene transfer vectors to develop anti-bacterial vaccines, and the ability of Ad vectors to code for single chain antibodies against specific antigens. The underlying concept is that Ad-based gene transfer vectors can be used to evoke systemic, robust acquired immunity, as well as rapid passive immunity against Y. pestis antigens, and that both forms of protection can be achieved with a single administration of a single vector. The proposal uses an in vivo gene transfer-based strategy with a single Ad vector to simultaneously evoke rapid humoral immunity against Y. pestis (via an anti-Y. pestis single chain antibody coded by the vector), while also functioning as a vaccine to evoke endogenous host responses against Y. pestis antigens (via Y. pestis antigens coded by the vector). The 3 specific aims outline studies to achieve these goals by developing the vaccine and single chain antibody strategies independently, and then combined. Aim 1. To evaluate the hypothesis that a vaccine based on a replication deficient Ad vector encoding the Y. pestis V and F1 antigens will evoke robust systemic humoral immunity against these antigens and protect against challenge with Y. pestis. Aim 2. To assess the hypothesis that an Ad vector encoding a single chain antibody against an epitope of Y. pestis V or F1 antigen will provide robust, rapid humoral immunity against these antigens and protect against challenge with Y. pestis. Aim 3. To examine the hypothesis that a combined passive and active anti-Y. pestis protection can be achieved with a single administration of a single Ad vector expressing anti-Y. pestis single chain antibody and Y. pestis antigens. [unreadable] [unreadable]
{ "pile_set_name": "NIH ExPorter" }
Experiments over the last three years have demonstrated that in the presynaptic terminal of the squid giant synapse the relation between inward calcium current and the synaptic release process is amenable to detailed examination using voltage clamp techniques. The actual relationship of calcium current to transmitter release will be done utilizing double voltage clamp experiments where the pre- and post-synaptic elements of the squid synapse are simultaneously voltage clamped. The purpose of these experiments is to determine the actual time courses of the presynaptic and pootsynaptic currents and thus to establish not only the nature of synaptic delay but, more important, the nature of ionic gain in synaptic transmission. Further study of the voltage-dependent calcium conductance change must be done to obtain information regarding the relationship between membrane potential and calcium current at varying extracellular calcium concentrations. A similar set of experiments will be done to determine the temperature dependence of this process (Q10) as well as on the time course of calcium permeability onset.
{ "pile_set_name": "NIH ExPorter" }
Description (Taken from the application): Somatic gene therapy may have the potential for treating diseases of bone that are resistant to current therapies. However, the cellular and molecular steps required to develop an integrated effective strategy are not fully developed. During the previous grant cycle, some of the problems were successfully resolved, including the ability to deliver a cell-specific expressed promoter-reporter construct within the context of a retrovector and the production of a pseudotyped retrovector capable of infecting a high proportion of primary marrow stromal cells without antibiotic selection. New models for assessing the effectiveness of bone marrow transplantation were developed using Collal-GFP transgenes whose expression is dependent on the stage of osteoblast differentiation. This proposal utilizes these tools to develop engineered marrow stromal cells that are enabled to engraft to bone, participate in bone turnover and produce a growth factor that will influence endogenous bone cells. We will confirm that viral transduced stromal cells can produce bone in vivo and we will develop a cell-specific promoter that can be activated in a controlled manner. The differentiation of stromal cells will be blocked with growth factors that expand the proportion of immature cells within the culture. The ability of these cells to participate in bone formation will be assessed by direct injection into the diaphyseal space of the femur and by tail vein injection into a murine model undergoing recovery from conditional ablation of the osteoblast lineage. The degree of engraftment, the ability to participate in new bone formation and the contribution to the self-renewing pool of osteoprogenitor cells will be determined based on the expression of GFP lineage markers of osteoblast differentiation of the donor cells. An additional aim will develop the steps required to make the host more receptive to engraftment. By analyzing each component of the strategy, it should be possible to develop a method for somatic gene therapy of bone in the mouse that may be applicable to man.
{ "pile_set_name": "NIH ExPorter" }
Using M-mode echocardiography, an estimate of left ventricular filling pressure in critically ill patients was calculated by determining the ration of the time from the onset of the QRS complex on EKG to mitral valve closure and the time from aortic valve closure to mitral value E-point. Using linear regression, an equation was derived which provided an estimate of pulmonary capillary wedge pressure. This estimate was compared to the pulmonary capillary wedge pressure measured by standard techniques using a pulmonary artery catheter, and a strong correlation was found. Serial studies in individual patients showed a good correlation of echo-determined pulmonary capillary wedge pressure with pulmonary artery catheter-determined pressure. Validation of this technique is ongoing and may allow some patients to be managed with a shorter duration of pulmonary artery catheterization as well as decreasing the need for repeated catheterization in some patients.
{ "pile_set_name": "NIH ExPorter" }
This project is a conceptual analysis to describe the normative assumptions necessary to translate empirical health services data into public policy. An analysis of this process for the development of public policy for cystic fibrosis newborn screening in Wisconsin has been completed. This analysis included several key findings. First, any benefits found during the research phase can not be assumed if a routine program dramatically alters the screening approach. Second, these benefits must be considered within the context of other potentially forgone benefits to patents with CF. Third, the benefits must be considered within the context of other potentially forgone benefits to other children. Finally, these benefits must be considered in contrast to potential harms to false positive families. A similar analysis of the process for cystic fibrosis carrier testing is being completed. Subsequently, an analysis to describe normative conditions under which such policies would be complementary or contradictory will be conducted.
{ "pile_set_name": "NIH ExPorter" }
The long-term objective of the proposed research is to understand the molecular mechanisms underlying vision in the fruitfly, Drosophila melanogaster. The goal of the current proposal is to understand the mechanisms regulating TRP, a novel plasma membrane Ca2+ channel specifically expressed in Drosophila photoreceptor cells. Changes in the concentration of intracellular Ca2+ are crucial for signal transduction in virtually every cell. During the last couple of years, it has been found that one of the most effective and prevalent Ca2+ influx pathways, referred to as capacitative Ca2+ entry, occurs via Ca2+ selective ion channels in the plasma membrane that are activated following depletion of intracellular Ca2+ stores. The mechanisms regulating these ion channels are poorly understood and the molecular identification of the proteins responsible for these plasma membrane Ca2+ conductances have been elusive. Based on a variety of electrophysiological, pharmacological and molecular analyses, it appears that TRP is the archetypical member of the family of capacitative Ca2+ entry channels. The approach to characterizing TRP takes advantage of a combination of molecular, biochemical, electrophysiological, pharmacological, genetic and germline transformation techniques. To study the mechanisms regulating TRP we plan to: 1) test the hypothesis that TRP function is regulated by interaction with calmodulin, 2) test the hypothesis that TRP localization to the base of the rhabdomeres is mediated by interaction with alpha-actinin, through the TRP ankyrin repeat, 3) test the hypothesis that TRP is regulated by interaction with GTP, 4) test whether the PEST signal is important for TRP function, 5) characterize the effects of systematic deletions in TRP on subunit assembly, activation and inactivation, and 6) characterize TRP expressed in the neural cell line, NG115-401L. The results of the proposed experiments should contribute not only to understanding visual physiology but to the widespread phenomenon of capacitative Ca2+ entry which has been proposed to be important in such diverse cellular processes as fertilization, cell growth, transformation, secretion, smooth muscle contraction, sensory perception and neuronal signaling. Several studies indicate that Ca2+ influx is important in memory involving NMDA receptors and other studies suggest that blockage of Ca2+ influx is important in controlling certain types of epileptic seizures.
{ "pile_set_name": "NIH ExPorter" }
The cornea is an attractive wound healing model because it has relatively simple and unique wound healing characteristics. The proposed studies will use type IV collagen purified fragments and chemically synthesized peptides with an objective of defining the molecular mechanisms involved in corneal epithelial cell adhesion and cell migration. The ultimate goal of these studies will be to develop peptide pharmaceuticals to modulate corneal wound healing. The information learned from these studies may have application to wound healing in general. In these studies we will evaluate the adhesion and spreading of corneal epithelial cells to type IV collagen, defined type IV collagen proteolytic fragments and synthetic peptides derived from type IV collagen. Next, we will evaluate the ability of chemically synthesized peptides from different regions of type IV collagen to stimulate the migration of corneal epithelial cells. In these studies both haptotaxis (migration to substratum attached) and chemotaxis (migration to solution phase ligands) will be determined. Integrins are a major class of cell surface receptors for extracellular matrix and basement membrane molecules. Integrins are complex heterodimers with important biological functions. We will define the integrin "receptors" on human corneal epithelial cells that mediate adhesion and migration to type IV collagen and specific synthetic peptide ligands derived from type IV collagen. a battery of specific anti- alpha chain antibodies and specific anti-beta chain antibodies will be used to delineate the full integrin complement. Lastly, we will test the influence of highly purified type IV collagen domains or fragments and chemically synthesized peptides derived from type IV collagen on the migration of rabbit corneal epithelial cells in the in vitro organ culture assay system. We have found three distinct synthetic peptides derived from the type IV collagen amino acid sequence that promote corneal cell adhesion: HEP-1 and HEP-2 from the NC1 region, and HEP-3 from the triple helical region. The HEP- 3 peptide, which promotes corneal epithelial cell adhesion, appears to represent a sequence of type IV collagen that is very important for stimulating cell migration. It seems likely that other sequences exist within the helical fragment in areas not yet tested that may also have cell adhesion or cell migration promoting capacity. These findings contribute to the molecular dissection of basement membrane structure and function, and they may serve as the basis for developing therapeutic agents to modify corneal epithelial behavior so as to have a salutary effect in problematic clinical conditions.
{ "pile_set_name": "NIH ExPorter" }
Research and experience document that caregivers of children with cancer encounter extraordinary stresses during the child's illness. These stresses are particularly severe during diagnosis and early therapy and can interfere significantly with a caregiver's ability to make reasoned and timely decisions on their child's behalf. With increasing survivor rates, it has become evident that decisions made in the early stages of cancer management can have profound long-term effects, adding to the distress caregivers feel trying to make the right decisions. Too help mothers of newly diagnosed children cope more effectively with these challenges, we conducted randomized controlled trials (R25CA65520, R01CA098954) to develop, field test, and evaluate the efficacy of the Bright IDEAS paradigm of problem-solving skills training (PSST), a cognitive-behavioral therapy shown to decrease anxiety and depression - two symptoms of post-traumatic stress commonly experienced among this group of mothers. Our findings clearly show that PSST significantly increases problem-solving skills (primary effect) and decreases negative affectivity (secondary effect) in mothers from a variety of racial, ethnic, and socioeconomic backgrounds. Particularly powerful and long-lasting effects were noted in Spanish-speaking mothers, an especially underserved population. In March 2010, the NCI designated Bright IDEAS as a Research-based Therapy/Intervention Program (RTIP) and entered it into the National Registry of Evidence-based Programs and Practices (NREPP). As an 8-session, in-person intervention, Bright IDEAS is labor intensive and, to date has only been available at a few institutions with trained personnel. However, as a part of the RTIPs evaluation process, the Dissemination Capability of Bright IDEAS was rated 5/5. This proposal is designed to meet the challenge inherent in this perfect score. We will also immediately broaden the scope of users by including fathers as eligible participants in this study of a new delivery paradigm we believe they will find appealing. Aim 1 is implementation of an engaging, easy-to-use on-line version of Bright IDEAS that would be available 24/7 to any person anywhere who has access to the Internet. Aim 2 is the use of Diffusion of Innovations theory to craft a framework for disseminating not only Bright IDEAS but also other similar interventions with the greatest effectiveness and efficiency. In past studies, we have shown that the human element (time and attention) inherent in in-person interventions is effective in bringing immediate relief of distress but not sufficient to maintain its benefits over time. In contrast, mothers receiving PSST increase their skills and continue to improve their sense of well being 3 months after the intervention. What we have not investigated is whether a computer-based intervention is as effective as (not inferior to) an in-person intervention. The results will have significant implications for future dissemination strategies, especially the use of Internet II and other emerging technologies.
{ "pile_set_name": "NIH ExPorter" }
York College's vision and mission statements describe it as an institution committed to academic excellence through teaching, learning, research and scholarship. One of the specific objectives in the college's 5-year plan, which is reflected in the plans of all academic departments, calls for creating an academic environment that values and encourages research and scholarship. The College envisions the MBRS/SCORE Program as a primary means of achieving this objective. The goals and objectives of the MBRS/SCORE Program are: 1) to strengthen the research infrastructure and change the College culture to support institutional research and research training by a series of activities resulting in: a) the appointment, promotion and tenuring of faculty in MBRS/SCORE biomedical disciplines, at least 75% of whom exhibit high potential for research; b) reduced workload for all new faculty in biomedical research disciplines concomitant with each faculty's development of a scholarly plan; c) at least 75% of faculty in MBRS/SCORE biomedical research disciplines participating in grantsmanship activities, 50% obtaining assistance in developing proposals, and 75% receiving start-up funding, d) all MBRS/SCORE faculty having adequate laboratory equipment, instrumentation and space; e) the York College-FDA Collaborations obtaining at least $1M in external funding for health-related research pursued by scientists from both institutions; 2) to maintain, and where possible, increase the participation of investigator-initiated research by a) increasing the number of biomedical research disciplines with MBRS/SCORE faculty from 6 to 7, b) increasing the percentage of MBRS/SCORE investigators by 25%; 3) to strengthen the research competitiveness of MBRS/SCORE faculty by a) facilitating them in meeting their respective career development plans, b) increasing their number of presentations at professional society meetings by 25%, c) increasing their number of peer-reviewed publications by 17%, increasing the number of faculty with competitive research programs i) by 29% in the S06 mechanism and by ii) 25% in R15 type mechanisms. To help achieve these goals and objectives, York requests support for the 5 faculty submitting MBRS/SCORE proposals: 4 Regulars and 1 Pilot. The Regular proposals come from biology (2), chemistry, and social work and the Pilot proposal comes from environmental health science. Two of these investigators are continuing S06 PIs, one is a current pilot investigator seeking Regular S06 funding, two are new investigators. The resources provided by MBRS/SCORE, will help the college continue to strengthen and diversify its research capability, research training capability, and continue to provide expanded opportunities for underrepresented minorities in biomedical research. [unreadable] [unreadable]
{ "pile_set_name": "NIH ExPorter" }
This subproject is one of many research subprojects utilizing the resources provided by a Center grant funded by NIH/NCRR. The subproject and investigator (PI) may have received primary funding from another NIH source, and thus could be represented in other CRISP entries. The institution listed is for the Center, which is not necessarily the institution for the investigator. We have several software packages which are distributed freely from our web site http://ncmi.bcm.tmc.edu. In addition to formal releases, daily snapshot versions are available for most packages. Available software includes EMAN, EMAN2, SAVR, AIRS and SAIL. AIRS is currently distributed as part of EMAN.
{ "pile_set_name": "NIH ExPorter" }
We propose to produce a 3D digital atlas of the C57BL6/J mouse brain by magnetic resonance microscopy (MRM). The atlas will be designed to serve as a general template for spatial mapping of all genetic information in the mouse brain and will be available to all molecular biologists. We will achieve 10 x 10 x 10 mum3 spatial resolution in the 3D MRM atlas through application of available technologies at the Center for In Vivo Microscopy and via development of new "active" proton staining technologies. The 3D MRM atlas will be available in at least three different endogenous and "active" proton stains. Further, a probabilistic (averaged from many brain specimens) atlas with error estimations will be created for registration of actual gene-expression data.
{ "pile_set_name": "NIH ExPorter" }