text
stringlengths 1
32.7k
| meta
dict |
---|---|
The major goal of this research is the development of mammalian culture systems to test for the mutagenicity of environmental agents. Compounds which are known mutagens are currently being examined for their action in several systems. Both the spontaneous and mutagen induced mutation rate is currently being examined at 3 loci in human lymphoid lines: the hpt locus specifying HGPRT activity; the as locus, specifying argininosuccinate synthetase activity, and the olocus, specifying ouabain resistance. Using murine myeloma cells lines spontaneous and mutagen induced changes in immunoglobulin biosynthesis are being investigated. Emphasis is being placed on the determination of the specific mode of action of mutagens in mammalian cells using two experimental approaches: 1) the structural changes in variant proteins are being determined and 2) systems are being developed which can be specifically reverted by mutagens of certain modes of action. All experiments using the mammalian cells are carried out in parallel in bacterial test systems both to verify the effectiveness of the mutagen and to determine the relationship between mutagenicity in simpler organisms and in more complex, differentiated mammalian cells. | {
"pile_set_name": "NIH ExPorter"
} |
Tuberculosis remains a serious and common disease worldwide. In the U.S. emergence of multi-antibiotic resistant strains, due to the rapid passage of Mycobacterium tuberculosis in patients with AIDS, poses a public health problem. Although the most frequently used vaccine in the world, there is no scientifically-based evidence that BCG prevents primary pulmonary tuberculosis and its protective effect against meningitis in children has not been related with a protective antigen or a host immune component. Our research into a new vaccine for tuberculosis is based upon the similarity of primary infection caused by M. tuberculosis with that of capsulated bacterial respiratory pathogens, viz: 1) epidemiologic data which show that tuberculous meningitis has a similar age distribution as capsulated bacteria; 2) the presence of a capsular polysaccharide on M. tuberculosis and other mycobacteria in vitro and in vivo; and 3) BCG and protein components of this and wild-type strains induce prolonged survival but do not protect animals against challenge with wild-type M. tuberculosis. We identified alpha(1 >2) glucose moiety in extracts of M. tuberculosis, but not in BCG, by its reactivity with pneumococcal type 12F typing antisera were not- productive. Almost all the polysaccharide extracted with saline from Mycobacteria was a glycogen-like molecule composed exclusively of D-glucose. Only a trace of this material precipitates with pneumococcal type 12F antisera and gives an identity reaction with dextran 1299 which has kojibiose residues. Conjugates prepared with kojipentaose did not elicit opsonophagocytic killing and did not protect mice against challenge with M. tuberculosis. Conjugates prepared with a conjugate of the glycogen-like molecule in two strains of M. tuberculosis induced opsonphagocytic killing and protected mice against challenge with wild-type M. tuberculosis. | {
"pile_set_name": "NIH ExPorter"
} |
Tissue structure and strength are the most relevant properties when assessing animal models related to musculoskeletal injury, disease and repair. Whether the treatment effects are created by genetic manipulation, metabolic challenge, simulated injury, surgical repair or other intervention, the net effects must be judged based on whether they have resulted in more or less tissue, whether that tissue has normal morphology, and whether the tissue or skeletal structure has increased or decreased mechanical properties. We have the necessary equipment and the demonstrated user expertise to assess the structural and mechanical properties of a range of musculoskeletal tissue and structures derived from animal models. There have been productive collaborations between investigators in the Research Base and the personnel of this Core, but these have been limited in number by the lack of a mechanism to support the acquisition and analysis of data by expert users and the training of non-expert users. The objective of this Musculoskeletal Structure and Strength Core (Core B) is to provide a mechanism to increase access to existing resources for x-ray based densitometry and imaging, and mechanical testing, and to thereby enable new interactions and enhance existing interactions between musculoskeletal researchers at Washington University. We will maintain protocols and equipment to promote quality control, provide technical support and training, and perform the following services on musculoskeletal structures and tissues from animal models (mouse to canine) generated by investigators in the Core Center Research Base. Aim 1: X-ray based imaging; available modalities include radiography, dual-energy x-ray absorptiometry (DXA), peripheral quantitative computed tomography (pQCT) and micro-computed tomography (microCT). Each of these is available for ex vivo or in vivo imaging. Aim 2: Mechanical testing;available modalities include whole-bone bending and compression, trabecular indentation, tensile testing of demineralized bone, tendon and tendon-bone insertion site tensile testing;muscle force measurement. By offering these Core services we will increase research productivity of established musculoskeletal investigators on our campus and facilitate non-musculoskeletal investigators wishing to bring new perspectives to studies in musculoskeletal biology and medicine. | {
"pile_set_name": "NIH ExPorter"
} |
The constant threats posed by Influenza and other RNA viruses of pandemic due to antigenic shift or transfer from an animal host, and of season-to-season variation due to antigenic drift can be most effectively countered by pervasive monitoring. Viral monitoring in the clinic and field would be assisted greatly by the availability of microfabricated devices capable of rapid, inexpensive genotyping. To accelerate the emergence of such devices we propose research to engineer and build two prototype microfabricated devices, one to detect influenza A sub-types, and the other to perform single nucleotide polymorphism (SNP) detection to monitor antigenic drift. We will also develop and integrate a purification system to prepare sufficient quantity and purity of RNA material from virus-containing clinical samples for the microfabricated device to perform reverse transcription-PCR (RT-PCR) and genotyping reactions. Specifically, we aim to Aim 1 - Develop a microfabricated device capable of producing viral RNA from biological samples with the levels of purity and concentration required to perform on-chip RT-PCR reactions. Aim 2 - On a microfabricated device, perform an RT-PCR reaction on HA 1 hemagglutinin domain of influenza A, producing double-stranded complementary DNA. Aim 3 - On a microfabricated device, perform restriction digestion reactions that can distinguish H1, H3, and H5 types of influenza A. Perform a multiplex reaction that can simultaneously detect RNA from influenza A and another RNA virus, such as influenza B or SARS. Aim 4 - On a microfabricated device, perform single-nucleotide polymorphisms that can distinguish antigenic drift in the HA1 domain of influenza A. Aim 5 - Integrate RT-PCR, restriction digestion, and electrophoretic separation onto a single microfabricated device, and integrate this with a system for RNA extraction from clinical, throat-culture, samples. | {
"pile_set_name": "NIH ExPorter"
} |
Encephalomyocarditis (EMC) virus induced diabetes mellitus in mice has similarities to a subset of juvenile onset diabetes in humans. Two otherwise similar variants derived from the M- strain of EMC virus are available for study that differ in their interferon inducing particle (ifp) phenotype and in their ability to cause diabetes in mice. The EMC-B variant is ifp+, produces high levels of circulating interferon (IFN) in infected mice, causes limited infection of pancreatic islets, and does not cause diabetes. When circulating IFN is neutralized by anti-IFN globulins, diabetes results from infection with EMC-B indicating that the interferon system, possibly the ifp phenotype, is a determinant of the diabetic outcome. The EMC-D variant is ifp-, produces lower levels of circulating IFN, destroys pancreatic islets and causes diabetes in mice. Key genetic differences must be responsible for the different biological properties of these closely related viruses. In order to determine the genetic basis for these properties we have cloned cDNA corresponding to the complete open reading frame of both variants. Their restriction endonuclease maps are identical for 12 different enzymes. Also, the first 103 nucleotides of the untranslated RNA at the 5' end of the genomes are identical. We propose to discover the genetic basis for the ifp phenotype and diabetogenicity of EMC virus by first comparing the complete nucleotide sequences of the B and D variants. The functional significance of differences in nucleotide sequence that are found to occur in the open reading frame will be ascertained by reference to the in vitro translation products of RNA transcripts and to the known functions of EMC viral proteins. Complete cDNA of the B and D variants will be synthesized for transfection of mammalian cells. With reference to differences in nucleotide sequence within or outside the open reading frame, chimeric molecules of B and D will be synthesized. The biological properties of the recombinant viruses will be determined in order to independently determine the genetic locus responsible for the ifp phenotype and diabetogenicity. This knowledge will further our understanding of virus virulence and the mechanism of interferon induction by viruses. Using data provided by this project, it may be possible to predict the virulence of human viruses from a knowledge of genetic structure. | {
"pile_set_name": "NIH ExPorter"
} |
This application is for a Research Scientist Development Award, Level I. The application describes a comprehensive program of training and research in the field of obesity and weight regulation. The proposed training in the areas of 1) regulation of food intake and body weight, 2) energy expenditure and clinical nutrition, and 3) body composition will contribute significantly to my scientific development and enable me to complete the proposed research more successfully. The research proposed in this application will examine three primary issues: 1) the long-term treatment of obesity in adults by very-low-calorie diet and behavior therapy; 2) the treatment of obesity in black adolescent females; and 3) the prevention of obesity in young children through a school-based intervention. The study on the treatment of obese adults will examine resting energy expenditure and body composition to determine if they are adversely affected by very-low-calorie diet (420 kcal/day). In particular, we will determine whether the prolonged use of a very-low-calorie diet reduces energy expenditure and lean body mass in a fashion that renders maintenance of weight loss more difficult. Changes in body composition will also be examined in the teenagers in the second study. It is important to demonstrate that youngsters do not lose vital lean body mass will dieting. This is the first major study of the treatment of obesity in black children. The school-based program for the prevention of obesity continues a line of investigation that our research team has been pursuing for the past several years. | {
"pile_set_name": "NIH ExPorter"
} |
Although new auditory evoked potential equipment was ordered late summer of 1987, delivery was not made until mid-April 1988. Dichotic studies using the digits test of Musick in normally aging and demented subjects did continue during that time (approximately 90 subjects were seen). Data collection is ongoing for the Dichotic Digits tests in these populations. Since mid-April of 1988, 26 subjects have been seen for gathering normative auditory brainstem response (ABR), data on our new auditory evoked potential equipment. Data is gathered at frequently used stimulus intensities, stimulus polarities, stimulus rate, and by age and sex. All of these conditions are studied using both circumaural and insert earphones. We are looking forward to also collecting data on insert electrodes should we be able to free up enough clinic time. It is essential that we continue to see as many normals as possible in order to be able to use our new auditory evoked potential equipment on our diverse patient populations as soon as possible. The insert earphones will provide even more comfortable testing conditions for many patients previously unable to tolerate heavy earphones or in whom myogenic artifact was a serious containment. Multi-frequency immittance norms will be collected as time permits. The software is still evolving and we anticipate data collection to begin in the fall of 1988. These studies will be helpful in evaluating the middle ear systems in patients with connective tissue disorders. It is pertinent to note that the Audilogy Clinic is primarily a service consultative facility, and the needs of the NIH patients always come first. Therefore, we see normal controls for this study only when there is no conflict with patient scheduling in Audiology. | {
"pile_set_name": "NIH ExPorter"
} |
The momentum to implement cervical cancer screening in low and middle income countries (LMIC) could be greatly enhanced if a robust point-of-care (POC) HPV test were available to utilize in the preferred screen- and-treat (SAT) approach. Furthermore, SAT programs could be made more attractive if HPV tests were modified, or additional tests added, to improve specificity of a single-round of screening without reducing sensitivity for the detection of cervical disease. This would reduce the magnitude of over-treatment without compromising the public health impact of the program. Our group at Columbia University has a well- established collaboration with the University of Cape Town, South Africa, with whom we have undertaken large clinical studies of cervical cancer prevention in low resource settings. We propose to partner with Cepheid, who have pioneered POC testing (GeneXpert) using self-contained real-time PCR cartridges and who are widely respected for their POC tests for tuberculosis that have become the global standard. Cepheid has recently developed a HPV POC test (Xpert HPV Assay) for use in Europe. We propose to re-engineer the Cepheid HPV test to make it more suitable for SAT in LMIC. The test will be re-engineered to improve specificity and positive predictive value (PPV) of HPV testing by: (1) utilizing differet cycle threshold (CT) cutoffs for different groupings of HPV genotypes, 2) investigating testing for only selected HPV genotypes and (3) investigating the potential of using quantitative HPV viral load measurements for HPV 16 and possibly other genotypes. To further address the challenge of over-treatment in the SAT approach, we propose to investigate the potential added value of tests for cellular mRNA cancer biomarkers. Cepheid has novel preliminary data demonstrating the utility of cellular mRNA biomarkers to distinguish high-grade cervical neoplasia. Some have already been formatted for the GeneXpert platform. These approaches are designed to improve the specificity and PPV of a single round of screening, and thereby reduce the number of women without cervical disease undergoing treatment, without seriously compromising sensitivity, so as to preserve the public health impact of a SAT program. | {
"pile_set_name": "NIH ExPorter"
} |
The purpose of the Baylor Child Health Research Career Development Award (CHRCDA) is to enhance the academic research and career development of junior faculty in pediatric medicine pursuing academic careers in translational research. We will continue to recruit bright, creative and productive young pediatric researchers from a variety of clinical and scientific backgrounds. The award will allow us to facilitate the development of their research by providing (a) salary support to ensure protected time, (b) research support, and (c) a structured program, with oversight of mentoring by an advisory committee and a didactic curriculum. The didactic curriculum will provide training in grant proposal writing, experimental design, and other areas necessary for a career in translational research. As the aim of the program is to train translational researchers to be able to compete for funding in their own right, during the first year in the program each Scholar will write a K-08 or K-23 grant proposal, and submit it to the NIH in the summer or fall of the second year. Each scholar is mentored through the program, with the mentor participating in many aspects of the program, including an annual Evaluation Retreat, where established physician scientists from around the country come to evaluate the program and review the scholars' research. Acceptance to the program is highly competitive, and is based on the quality of a research proposal developed by the applicant with the help of his or her mentor, the quality of the mentor, and the committee's assessment of the applicant's ability to succeed in the program, write peer-reviewed publications and obtain independent research funding. All applications will be reviewed by the Program Advisory Committee of eight outstanding faculty members, and we anticipate supporting four scholars each year. Over the last 16 years, the CHRCDA has fostered the career development of 53 physician scientists, 36 of whom are current members of the Baylor faculty and another nine of whom are academic pediatricians in other institutions. The program has been highly successful in guiding the research and career development of junior faculty at Baylor College of Medicine. Overall, the CHRCDA has achieved its original goal of fostering research for the benefit of the health of children and is a critical component of the faculty development program in the Department of Pediatrics. This grant proposal seeks to continue this successful program. [unreadable] [unreadable] [unreadable] | {
"pile_set_name": "NIH ExPorter"
} |
Hematopoietic stem cell transplantation (HSCT) is a successful treatment option for many patients with severe hematologic diseases. However, large numbers of transplantable cells are needed and patient survival is compromised when donor hematopoietic stem cell (HSC) numbers are limited. This is the case when umbilical cord blood (CB) is utilized as a donor source for transplantation into adults. Given that CB is readily available, has a lower histocompatibility requirement, and carries a reduced risk of graft vs. host disease, there are clear reasons for using CB for HSCT. To overcome the problem of limited cell numbers, cytokines and growth factors are being utilized to facilitate ex vivo CB expansion pre-transplantation or as therapy post- transplant. The effects of cytokine treatment on stem cell trafficking during transplantation have not been described. Our long-term goals are to understand the mechanisms that govern HSC trafficking and to design innovative methodologies to improve the effectiveness of HSCT. Achieving these goals would have a dramatic impact on outcomes for adult patients undergoing cord blood HSCT. Towards this goal, we recently demonstrated that inhibition or loss of CD26 (DPPIV/dipeptidylpeptidase IV) significantly improved HSCT efficiency. We hypothesize that granulocyte colony-stimulating factor (G-CSF) and granulocyte macrophage-colony stimulating growth factor (GM-CSF) treatment up-regulates the peptidase CD26 resulting in decreased HSCT efficiency and that CD26 inhibition can reverse this negative effect. To investigate this, we propose: Aim 1) to evaluate the in vitro effects of G-CSF, GM-CSF and stem cell factor (SCF; also called steel factor or kit ligand) on human cord blood hematopoietic stem and progenitor cell (HSC/HPC) function. We will do this by evaluating cytokine induced changes in 1a) CD26 RNA expression, CD26 protein expression, and CD26 peptidase activity, and 1b) cell migration, cell adhesion, and progenitor colony formation in the presence or absence of CD26 inhibitors. Aim 2) to evaluate the in vivo effects of G-SCF, GM-CSF, and SCF on human cord blood HSCT efficiency and the use of CD26 inhibition to overcome any detrimental effects that result from cytokine treatment. We will do this by measuring cytokine induced changes in 2a) the homing of human donor HSC/HPC to the bone marrow of NOD/SCID/B2mnull immunodeficient recipient mice 24 hours post transplant, and 2b) the engraftment of short term repopulating cells of myeloid restricted (STRC-M) and dual myeloid-lymphoid (STRC-ML) lineage as well as long term repopulating cells (LTRC) into NOD/SCID/B2mnull immunodeficient mice at 3, 6, and 12 weeks post transplant. The involvement of CD26 in mediating any negative effects resulting from prior cytokine treatment will be directly assessed by treatment of the donor cells with CD26 inhibitors. [unreadable] [unreadable] Project Narrative Relevance: The purpose of this proposal is to explore a new direction in the field of stem cell transplantation by developing an understanding of the negative effects of cytokine usage on cord blood hematopoietic stem and progenitor cell trafficking, to determine whether these effects result mechanistically from an up-regulation of CD26, and to evaluate the potential novel use CD26 inhibitors to mitigate this effect. The clinical implications of this proposal are extremely high with respect to adult hematopoietic stem cell transplant patients who are currently unable to receive cord blood as a donor cell source due to limited numbers of stem cells obtainable from a single cord blood collection. We expect upon successful completion of the "proof of principle" style aims proposed here that a novel CD26 inhibitor based treatment strategy will emerge for use in combination with cytokine based therapies. Furthermore, we anticipate that with the appropriate subsequent research and clinical trials this research will have a positive impact on the survival of cord blood stem cell transplant patients. [unreadable] [unreadable] [unreadable] | {
"pile_set_name": "NIH ExPorter"
} |
ABSTRACT Alzheimer's disease (AD), the most common form of dementia, is a crippling neurodegenerative disease that is growing quickly in prevalence. Currently, there are no FDA approved medications that serve to prevent or reduce the pathology; all drugs are merely cognitive enhancers used to offset the deficits of dementia. Genome-wide association studies (GWAS) have been conducted to supplant the already acquired knowledge surrounding AD pathology and have recently identified one of several genes, Triggering Receptor Expressed on Myeloid Cells 2 (TREM2), as a genetic node in the risk of developing AD. The main function of TREM2 significant to AD is stimulation of macrophage and neutrophil-mediated inflammatory responses, suggesting the exacerbation or alleviation of TREM2 to AD pathology is heavily based on chronic, injurious neuroinflammation as well as amyloid-? clearance. Several scientific studies published within the last five years have produced mixed results as to whether decreasing the activity of TREM2 with antibody antagonism or gene knockout mitigates or provokes the pathology of AD. Therefore, further validation of these findings is warranted, particularly pertaining to modulation of TREM2-TYROBP signaling to assess the effect of increasing phagocytosis of amyloid-? oligomers and fibrils. In order to pursue this topic, both agonistic and antagonistic small molecules were screened for and characterized for their potential to modulate TREM2-TYROBP signaling in vitro. Using an in-house developed luciferase assay validated as a robust method for assessing TREM2-TYROBP signaling in Human Embryonic Kidney cells (HEK293) transfected with a construct of both genes, we identified several hits as agonists or antagonists of TREM2-TYROBP coupling. Our next aim is to validate these hits in BV2 murine microglial cells and primary cultured murine microglia. After further characterization of these compounds in vitro, they will be tested in vivo for toxicology, bioavailability, and therapeutic efficacy in a knock-in mouse model of AD using APPNL-G-F knock-in mice. Therapeutic efficacy will be evaluated in consideration of reduced amyloid burden and reduced age-related cognitive decline as assessed with biochemistry, immunohistochemistry and behavioral examinations, respectively. Successful completion of these studies will 1) contribute to the understanding of TREM2 and its in role AD pathology and 2) present highly validated compounds as tools for further research in this area. | {
"pile_set_name": "NIH ExPorter"
} |
The eukaryotic metaphase chromosome represents an approximately 10 to the 4th power-fold condensation of DNA. This condensation is probably achieved by several orders of DNA packing, each order dependent on unique DNA and protein interactions. The relationship of the nature of DNA packaging to cytological chromosome analysis techniques (i.e., G-banding) has not been established. The goal of this research is to characterize the interactions which stabilize DNA-folding in metaphase chromosomes and relate this to the banding patterns observed in the analysis of human metaphase chromosomes. An understanding of metaphase chromosome structure may lead to more detailed analysis of human chromosomes by the standard techniques, as well as new methods of analysis based directly on chromosomal protein content. The strategy of this research is to combine defined molecular probes of alterations with cytological observations. A portion of the work will deal with probes of isolated chromosomes in solution followed by biochemical and cytological analysis. In addition, antibodies to non-histone chromosomal proteins will be used to investigate the distribution of these proteins on metaphase chromosomes. | {
"pile_set_name": "NIH ExPorter"
} |
To explain how capillaries form, as well as the organization of fibroblasts and keratinocytes in response to cutaneous injury, recent emphasis has been placed on the extracellular matrix and its ability to control morphogenic processes. It is now recognized that cells are capable of integrating both chemical and mechanical signals from the matrix which directly influences the expression of differentiation and growth specific genes. Cell locomotion, as an essential component of endothelial and mesenchymal repair and regenerative processes, requires the coordinated interaction between cell surface adhesion receptors and specific extracellular matrix (ECM) molecules. Therefore, identification of molecules which regulate the process of cell migration is important for the development of new targeted therapies for accelerating impaired dermal wound healing or enhancing repair processes in the high-risk wound. Fundamentally, the pericellular composition and concentration of surrounding matrix components has a direct effect on cell speed, in part, by controlling the adhesive strength of the cell-substrate interaction through the level and availability of bound legends for cell adhesion receptors. Typically, low cell speed occurs on ligand densities which yield either very weak or extremely strong levels f adhesive strength. Within this framework both the levels of specific glycoprotein and proteoglycan synthesis, as well as their degradation by proteases or other factors will influence adhesive ligand availability. However, mediation of these effects requires ligand interaction with specialized adhesive contacts on the cell surface which integrate and coordinate the environmental signals responsible for cell movement. In addition to containing integrin receptors, a recently cloned transmembrane heparin sulphate proteoglycan (HSPG), syndecan-4, has also been localized to these adhesive complexes. Specific Aim 1: Define the role of syndecan-4 in modulating cell migration in relation to substrate ligand density and its correspondence with effects on overall cell/substrate adhesiveness. Specific Aim 2: Characterize the spatiotemporal expression pattern of syndecan-4 in migrating keratinocytes, dermal endothelial cells, and fibroblasts, as well as in a model of cutaneous injury. | {
"pile_set_name": "NIH ExPorter"
} |
Project Summary The goal of this proposal is to increase our understanding of the therapeutic benefits of using platelets as delivery systems for disease treatments, with the long-term goal of developing platelet-based therapeutic cell treatments for metabolic diseases, specifically, lysosomal storage diseases (LSDs). LSDs are inherited metabolic diseases that are characterized by an abnormal buildup of various toxic materials in the body?s cells as a result of lysosomal enzyme deficiencies. These malfunctioning enzymes represent a group of about 50 different genetic diseases and, though individually rare, their combined prevalence is estimated to be 1 in every 8,000 births. LSDs affect different parts of the body including the skeleton, brain, skin, heart, and central nervous system. All patients with LSD have a limited life expectancy. Platelets are anucleate blood cells that circulate throughout the body and play an important role in homeostasis, wound healing, angiogenesis, inflammation, and clot formation. Platelets are naturally filled with secretory granules that store large amounts of proteins, which are formed from the cytoplasm of megakaryocytes (MKs), their precursor cells. When platelets are activated, a large number of bioactive proteins are released from their granules to participate in a myriad of physiological processes. We propose to take advantage of platelets? innate storage, trafficking, and release capacities, to engineer them as delivery vehicles for the development of next generation delivery methods for lysosomal enzymes to treat patients with LSDs. My central vision is to engineer blood platelets to control the secretion of enzymes required for proper lysosomal function as a therapeutic treatment for patients with LSDs. I will build upon my previous work where I have developed novel genetic tools and circuits to re-engineer cells to perform specific tasks to systematically design, build, and characterize new genetic tools for packaging lysosomal enzymes into platelets. Additionally, I will rationally design receptors that are capable of activating platelets to trigger the release of enzymes upon binding to specific drugs and/or binding to tissue specific peptides. Success from this proposal will result in paradigm-shifting therapies with unprecedented levels of flexibility, precision, and personalization. The resulting engineered platelets will be the most sophisticated therapeutic agents ever developed for treating metabolic disorders. Furthermore, the genetic tools and design principles developed here will serve as a general platform that can be combined with any other treatments to complement existing therapies, thus this work will have an immediate and broad impact on many metabolic disorders. | {
"pile_set_name": "NIH ExPorter"
} |
Technical Assistance for Cancer Registrar Tasks | {
"pile_set_name": "NIH ExPorter"
} |
Spoken language is a fundamental skill acquired during the first three years of life. However, a number of young children have language problems that begin to emerge during these years. Research has suggested that delays in language acquisition are associated with a number of negative social and academic outcomes (Fujiki et al., 1996; Rescorla, 2002), including 'acting-out' behaviors, which are commonly referred to as externalizing problems (Redmond & Rice, 1998). Further, both externalizing symptoms and language have been associated with lower levels of parental sensitivity and higher levels of parental rejection. However, little research has examined the role of parenting in the association between language delays and co-occurring externalizing symptoms. The proposed research was designed to explore the development of language problems and the association between emerging externalizing symptoms and language problems in a sample of low-income, high-risk children who were followed from the ages of 2 to 5. Specifically, the project will: 1) Explore trajectories of early language development including transient versus more permanent delays; 2) Examine reciprocal associations between language problems and externalizing symptoms from ages 2 to 5; 3) Study the moderating role of parenting in attenuating reciprocal effects between language and externalizing problems in early childhood. Group-based trajectory modeling, autoregressive structural equation modeling and multiple regressions will be the statistical tools used to explore these relationships. The findings of the proposed research should inform educators, clinicians and researchers with a more comprehensive picture of the early development of language problems, and associations between emerging externalizing symptoms and language problems. Further, if parenting is found to moderate associations between language and externalizing problems, this knowledge could lead to potential targets for future intervention. For example, if a child is displaying language problems, it may be possible for clinicians to intervene at the level of parenting as well as targeting language. Addressing both problems simultaneously may improve a child's language and decrease their risk for later externalizing problems. [unreadable] [unreadable] [unreadable] | {
"pile_set_name": "NIH ExPorter"
} |
Although much is known about client prognostic indicators of substance abuse treatment outcome, less is known about how treatment impacts outcome. In particular, there is a need for better information on how to allocate inpatient and outpatient care in relation to the severity of substance abuse, psychiatric disorders and life context factors. Such research holds promise for improving the effectiveness and reducing the overall costs of substance abuse treatment. The primary aims of this project are to: (1) assess the cost-effectiveness of residential and outpatient treatment for substance abuse; (2) document treatment and posttreatment service utilization; (3) evaluate three theory-based patient-treatment matching hypotheses; and (4) assess the patient- treatment matching utility of an established patient assignment system. A total of 600 participants with drug abuse problems will be randomly assigned to residential treatment (N = 200), outpatient treatment (N = 200), or to a usual care group in which participants receive a program placement based on the American Society of Addiction Medicine's patient assignment criteria, followed by treatment in a flexible continuum of care (N = 200). These clients will be assessed on a wide variety of health and psychosocial measures prior to treatment and at six- and twelve-months after discharge from treatment. Service utilization and cost data, as well as data on treatment participation, intermediate outcomes, and life context factors, will also be collected and analyzed. The proposed study will overcome the weaknesses of previous studies in several ways: (1) study clients will be randomly assigned to alternative treatment settings; (2) multiple outcomes will be measured in a longitudinal design and substance use will be verified using urine screens and collateral reports; (3) services utilization will be assessed using a comprehensive standard service taxonomy and individual client costs will be determined using valid and reliable methods; and (4) multivariate analyses will be undertaken to examine the processes of treatment and to test a priori hypotheses concerning differential effects of treatment setting for different types of clients. The cost and outcome data will be analyzed using a number of multivariate statistical analyses, including the Theta methodology, which is designed for analyzing outcomes and costs together. Structural equation modeling, regression, and analysis of covariance will also be used in cost- effectiveness analyses. Comparisons of treatment outcomes and tests of matching hypotheses will be conducted using selection modeling, analysis of covariance, and classification and regression tree analyses. Mediational models of treatment processes that link treatment characteristics to intermediate outcomes and final outcomes will be assessed using a path analytic strategy. | {
"pile_set_name": "NIH ExPorter"
} |
This proposed research will establish a cohort of workers exposed occupationally in the past to dimethylnitrosamine. Prospective observation of their full mortality experience, from the time of first exposure, will be conducted. Expected and observed rates of death by cause will be tabulated to ascertain if unusual mortality patterns exist. Such data will have importance in assessing the potential human carcinogenicity of dimethylnitrosamine and in assessing the potential risk of environmental exposure of N-nitroso compounds. | {
"pile_set_name": "NIH ExPorter"
} |
The Interinstitute Genetics Program and the Genetics Clinic supported by the Clinical Center offer a multidisciplinary approach to patients with genetic disease (Z01 CP 05139-06 CEB). Researchers from all Institutes are involved in the program Patients evaluated in the clinic represent a broad spectrum of genetic diseases. During the past year, the approximately 200 persons seen represented about 60 distinct disease categories. Because of the high frequency of ocular involvement in many of the cases, almost all the patients were evaluated by Clinical Branch staff or were discussed in consultation. The clinic serves as a source of interesting case material concerning patients with inherited or developmental abnormalities of the visual system. | {
"pile_set_name": "NIH ExPorter"
} |
SUMMARY: The overall goals of this project are to define the host cell and virus functions responsible for the regulation expression of viral functions during polyoma virus replication. In particular, the control of the switch-over to the expression of late gene functions following the onset of viral DNA synthesis is under study. A unique new type of polyoma variant (3049) has been described whose phenotype is recognized by the production of capsid proteins at 2-3 fold the rate of that found in a wild-type virus. This increased rate of capsid protein synthesis is correlated directly with the presence of 2 fold more virus-specific, polyadenylated RNA both in the nucleus and on cytoplasmic polyribosomes. This finding has suggested that the phenotype of the variant virus is due to an altered rate of transcription of at least the late genes coding capsid protein. BIBLIOGRAPHIC REFERENCES: Tachovsky, T. G. and Hare, J. D., A polyoma virus strain with enhanced synthesis of capsid protein. J. Virology 16, 116, 1975. Hare, J. D., Distinctive alterations of nucleoside, sugar and amino acid uptake by sulfhydryl reagents in cultured cells. Arch. of Biochem. and Biopsy. 170, 347, 1975. | {
"pile_set_name": "NIH ExPorter"
} |
Despite the improved activity of combination antiretroviral therapies in patients with HIV infection, the antiviral, immunologic, and clinical impact of such therapies appears to be incomplete and is often transient. In addition, treatment with the agents noted above may be associated with significant drug interactions and side effects. Combination therapies with complicated administration regimens also pose a challenge to some populations requiring antiretroviral therapy. Therefore, the identification of novel and conveniently administered agents in combination strategies for treating patients with HIV infection would be of great benefit. Adefovir dipivoxil, a nucleotide reverse transcriptase inhibitor, has been shown to be effective in vivo against HIV by its ability to reduce viral load measurements including p24Ag and HIV RNA. Adefovir dipivoxil at a dose of 60 mg daily has been utilized in previous trials but only as a dose reduction from 120 mg in response to clinically significant toxicities or intolerance. Previous clinical trials have demonstrated a clear dose-dependent relationship for adefovir dipivoxil related adverse events. However, no clear dose dependence has been established for the antiviral activity of adefovir dipivoxil at the doses clinically studied: 125 mg to 500 mg daily. The antiviral activity or safety profile of adefovir dipivoxil 60 mg daily has not been examined. It is possible that a daily dose of 60 mg has the same anti-HIV effect as 125 mg. Therefore, the rationale for conducting this protocol is to determine if there is antiviral activity of adefovir dipivoxil at a lower dose than those tested previously in human trials. | {
"pile_set_name": "NIH ExPorter"
} |
Surgeons are required to master complex and ever advancing knowledge and skills. Given both technical advancements and operational changes in the field of surgery, surgical mastery is becoming a task that requires more learning in less time. Thus, there is great need to understand ways in which surgical education and training can become more efficient. New mandates by the Accreditation Council for Graduate Medical Education's (ACGME) Next Accreditation System call for continuous monitoring of performance outcomes based on competency of essential tasks for clinical practice. This mandate sets a high bar for assessment and documentation of competency. The current proposal focuses on the development and validation of an assessment system for surgeons: the automated performance assessment system (APAS). The goal of APAS is to provide automated and objective feedback to its users. Specifically, it is an assessment that utilizes two previously validated technologies (open surgery simulators and motion sensor tracking) to provide feedback on a number of performance variables. The purpose of this research is twofold: (1) to capture data in order to best understand what variables are associated with surgical mastery, thus creating assessment criteria for APAS, and (2) Validate APAS as a reliable measurement tool for competency and mastery skills assessment for open surgical tasks. Both aims will be completed through between-groups experimental designs, where the experience level of the participant indicates the group and serves as the independent variable. The product of this research will be an assessment tool that is the first of its kind, evaluating both decision making and technical skills. This research proposes an end product that will change the nature of surgical training, enabling more timely, systematic, and detailed assessment. The goal of this research, innovation in medical training in the field of surgery, is one shared by the University o Wisconsin. As such, the applicant will be supported throughout this project both through access to mentors who themselves are leaders in the field of Surgery Education and Education and Technology. In the two year timeline of this project, this applicant will complete the research to produce a new form of surgical assessment, and complete the coursework to obtain a Master's degree in education. These experiences will serve as the first steps towards becoming a leader in the field of surgical education. | {
"pile_set_name": "NIH ExPorter"
} |
Most research to date using animal models of drug abuse has focused on the effects of psychostimulants on brain neurochemistry and behavior in adult, prenatal, or preweanling animals. There has been little research focusing on the effects of psychostimulants on adolescent rats. However, it is well known that a large number of people who use drugs started as teenagers. Because of this, it is of interest to examine the effects of psychostimulants in this adolescent population in order to determine whether there may be a difference in how this class of drugs effects this age cohort, and on how drug use at this age affects adult susceptibility to addiction. The specific hypothesis of this proposal is that psychostimulants have different effects on the behavior of periadolescent rats than of adult rats, and that exposure to drugs during this critical phase may alter the neurochemical underpinnings of stimulant abuse as an adult. A number of studies have examined the effects of pre-exposure to drugs on later response to cocaine. In all of these studies, both the pre- and post-exposure phases were conducted after the animal had reached adulthood. These studies have shown that pre-exposure to a number of different drugs can influence both the neurochemical and behavioral effects of subsequently administered psychostimulants. Repeated exposure to direct or indirect dopamine agonists in adult rats produces sensitization to the behavioral effects of psychostimulants. Our preliminary data show that, in adult rats, sensitization is evident both during a seven day treatment period, and 10 days after the treatment ends. In contrast, no sensitization is apparent in periadolescent rats treated under the same drug regimen. The aim of these experiments is to compare the effects of several drugs during the periadolescent phase to during the adult phase. Rats will be treated daily for one week with either cocaine, methylphenidate (ritalin), MDMA (ecstasy) or the appropriate vehicle during the periadolescent or adult phase. Locomotor activity will be tested for one hour daily during this period. Immediately subsequent to this period, or one month later, markers of dopamine and serotonin neurochemistry will be measured. In addition, one month later, the response to cocaine will be tested again on locomotor activity. An understanding of the differential effects of drugs during the periadolescent phase and of how this use impacts drug effects in adults may lead to different treatments for different age groups, as well as a better understanding of how drug use in adolescence may lead to psychostimulant abuse in adulthood. | {
"pile_set_name": "NIH ExPorter"
} |
Abstract Aim 1: Implement the NN/LM Document Delivery and Interlibrary Loan Plan in the Region. The NN/LM MCR will support programs to provide biomedical and health information to affiliated and unaffiliated health professionals, researchers, librarians, and the public. Aim 2. Efficiently and effectively support access to biomedical and health information. The NN/LM MCR will identify, disseminate, and promote effective practices as described by our Network members. Aim 3. Recruit members to participate in MedPrint. The NN/LM MCR will identify potential Network members for the Medical Serials Print Retention program and attempt to recruit their participation. Aim 4. Support Biomedical big data. The NN/LM MCR will begin a program to increase our Network members' knowledge and skills as related to biomedical big data. We will also collaborate to develop a health big data specialization and symposium on clinical data. | {
"pile_set_name": "NIH ExPorter"
} |
1\\J54 Interdisciplinary Researchoh Stress, Self-Control &Addiction Principal Investigator/Program Director(Last, First, Middle): SINHA, Rajita DESCRIPTION: See instructions. State the application's broad, long-term objectives arid specific aims, making reference to the health relatedriess of the project (i.e., relevanceto themission of'theAgency). Describe concisely the researchdesign arid methods for achievingthesegoals.Describe .the rationale and techniques you will use to pursue thesegoals. . . . : In addition, in two or three sentences,describe in plain, lay languagethe relevance of this research to public health. If the application is funded, this description, as is, AWN become public information: therefore, do not include prbprietary/cohfidehtialinformation. DO NOT EXCEEDTHESPACE ''' Tobacco smdking, alcohol cbhsurhption and bverconsumptibn of rich and high fat 'comfort'foods are the top three causes^of preventable death and disease in the US today. The persistent and compulsive engagement in th'ese addictions despite serious health, social and legal consequences is a common feature- Emerging data indicate that self control mechanisms ate critical in perpetuating this compulsive engagement in addictive behaviors^Stress, which plays a key role in addiction, other psychiatric illnesses and in many chronic diseases, alsb facilitates lapses in self control. Although researchoh the links between stressand addiction, stress and psychiatric disorders and stress and chronic diseasesexists, systematicresearcho'ri the intetactidris between Stress, self control and addictibn have been rare. This three-wayinterplay between Stress;: selfcbntrbl and addictibri is complex^arid requires ah interdisciplinary (ID) conceptual ffarhewbrkwith cbflabtiratiye telffi^-based approaches;bfstOdy that include the rhuitiple brain, bbdy, behavioral and sbciai syslerhSVWe therefore^ stress, self control and addiction GSSA) tfiat brings^togetherbver 50 leading'scientists;who conduct research relevant to a nurhber of Nlfi slitutes^MH^lA, NlDArNlAAA, NHLBI/NGI;NfGHD, NllDidK, NlEHS, NINDS, NIDtD). These experts rephSseritirigf20 disciplines that spah 5 schools (IvJedicihe, Arts arid Sciences, Mahagerheht, Nursihg ahd / public Heaith) arid three academic Institutions will cbllaborate as a team to: (A) identify Mechanisms iiftd&riyirici the development 6f stress-related effectson self-controlin the addictive behaviors of smc-kihg, dnnkihgand overeating;(B) evaluate self-control mechanisms in thepathophysiology of chronic stress arid addictionf and (0) develdp social, behavioral andpharmacological strategies to increase self-control and ^cWa'setri^se^a^aic^ve behaviors, these gbais will be acCbmplishedusing animal mbdels (hbn-hutnan pfirfllte;arid fbdetits) iahd hurhdn studies that include Children and adults. Achieving these gbals v\/iil have a direct irtipact in reducing rnbrbidity atVd mbrtaJityresulting from each of these disbrderS.Thei gb[unreadable]is will be '/ achifeiyed by addriessing the following specific aims: (1) to establish an interdisciplinary research cotisortiurh by brihgifitj tbgetheYleadihg biological, behavibfal and sbciai scientists to examine the mechahisrfis underlying ^tressvself-cbrttrol and addictive behaviors;(2) To conduct programmatic, team-based collaborative research tbunderstarid the prbcesses underlying stress and self control that promote and maihtaihcbrripulsive Srhokirtg, drinking artd bvereatihg;(3) To develop new sbciai, behavioral and pharmacological pfeventibh and treatment strategies to decrease stress, increase self-cbritrbl and prevent and decrease addictive behavibrs;(4) to foster career development and mentoring of interested students ahd scientists from varied disciplines in cbllabbrative, interdisciplinary research on stress, self control, addiction and related chronic diseases;and (5) To disseminate research findings to professional audiences arid the pubiic, including partnering with the Community to Collaboratively address emerging social( health pbiicy and bioethical issues raised by studies of stress, self-control and addictive behaviors. PERFORMANCE SltE(S) (organization, city, state) Yale University, New Haven, CT University of California- Irvihe, Irvine, CA Flbrida State University, Tallahassee, FLA PHS 398(Rev. 04/06) Page ^ FormPage2 1:U54 Interdisciplinary Research on Stress, Seif-CoritrdI &Addiction Principal investigator/Program Director (Last. First, Middle): SINHA, Rajita KEY PERSONNEL. See instructions. Use continuation pages as needed to provide the required information in the format shown below. Start with Principal Investigator. List all other key personnel in alphabetical order, last name first. Naftie eRA Commons User Name Organization Role oh Project SEEATTACHED OTHER SIGNIFICANT CONTRIBUTORS Name Organization Role on Project SEE ATTACHED Huifiah EnibryOihic Sfeifi Ceils |^ No D Yes If the proposed project Involves human embryonic stem cells, list below the registration number of the specific cell llne(s) from the following list http://^tehiCeHs.nih.qbv/rieqistfV/index:siSp. Usedohtin'dation pages ashee'ded. If a specific line cannot berefe'rehceclatthis time, include a statement that one from the Registry Will be'.used,^, .[unreadable]'.-.[unreadable],'... .: . . ., '[unreadable]...[unreadable]:. [unreadable]:[unreadable].:. Cell Line Disclosure Permission Statement Applicable to SBIR/STTR Only. SeeSBIR/STTR instructions. [U Ye^ Q No PHS398 (Rev. 04/06) ~~ ~ Page 3 ."..'.''.'Form Page 2 NurtiberWefo/tovvrhgpages corisecutiveiy throughout (he application. Do not use suffixes such as 4a, 4b. : U54 Key Faculty in the Yale Interdisciplinary Research Consortium NIH Roadmap Application on Stress, Self Control and Addiction (IRCSSA) Name eRA Commons Organization Project Role Sinha, Rajita PhD RAJITA geqrge_m_ander Anderson, George PhD son Arnsten, Amy PhD AMY_ARNSTEN Axelrod, Seth PhD SAXELROD Baumeister, Roy PhD BAUMEISTER Blumberg, Hilary PhD HPBLUMBERG Bockenstedt, Linda MD bockenlk Brandt, Cynthia MD CBRANDT Brownell, Kelly PhD KBROWNELL Carson, Richard PhD RECARSON Cleary, Paul PhD PDCLEARY Constable, Todd PhD constable12 DiLeone, Ralph PhD dileone Ding, Yu-Shin PhD YU_SHIN_DING Duman Duman, Ronald PhD Falba, Tracey PhD TFALBA Fletcher, Jason PhD JMFLETCHER Frost, James PhD JJFROST Yale University Yale University Yale University Yale University Florida State University Yale University Yale University Yale University Yale University Yale University Yale University Yale University Yale University Yale University Yale University Duke University Yale University Yale University Project Director;Principal Investigator PI: Project 14;Investigator: Projects 8, 10 PI: Project 2;Investigator: Projects 4, 5, 6, 9 Investigator: Projects 1 (U54), 12 (R25); Professional Project Manager PI: Project 10;Investigator: Projects 4, 11, 13 PI: Project 6;Investigator: Projects 4, 5, 7, 8 Co-l: Project 1 2 (R25) Co-l: Project 13 IRC Steering Committee; Consulting Investigator :Project 1 (U54), Project 12 (R25) Consulting Investigator : Project 8 Co-l: Project 11 Consulting Investigator: Project 7, Project 13 (P30);Collaborator: Project 6 Co-Pi: Project 3 Investigator: Project 8 IRC Steering Committee; Consulting Investigator: Project 1 (U54) Co-Investigator: Project 1 1 .Co-Investigator: Project 1 1 Consulting Investigator: Project 8 PHS 398/2590 (Rev. 09/04, Reissued4/2006) Page 4 Continuation Format Page Fulbright, Robert MD RFULBRIGHT Gallo, William PhD WGALLO Gelernter, Joel MD jGELERNTER Goldberg, Steven PhD Goldbergs Gray, Jeremy PhD jgrayl Green, Donald PhD GREEND Grey, Margaret DrPH MMGREY Grilo, Carlos PhD cgrilo Hohmann, Andrea PhD AHOHMANN Horvath, Tamas PhD THORVATH DVM Kalmar, Jessica PhD JKALMAR Katz. Stuart MD SDKATZ Kaufman, Joan PhD joan r. kaufman Keenan, Patricia PhD PKEENAN Kim-Cohen, Julia PhD KIM-COHENJ Yale University Yale University Yale University NIDA Yale University Yale University Yale University Yale University University of Georgia Yale University Yale University Yale University Yale University Yale University Yale University Krystal, John MD jkrystal Yale University Lampert, Rachel PhD LAMPERT Yale University Landwirth, Julius JD MD JLANDWIRTH Yale University Lee, Daeyeol PhD DAEYEOLLEE Yale University PHS 398/2590 (Rev. 09/04, Reissued 4/2006) Page 5 Consulting Investigator: Project 7 Co-Investigator: Project 11 Co-Investigator: Project 14 (P30); Consulting Investigator: Project 1 (U54) Consulting Investigator: Project 5 Co- 1: Project 13 (P30);Investigator: Project 7 IRC Steering Committee;Consulting Investigator: Project 1 (U54) IRC Steering Committee; Consulting Investigator: Project 1 (U54) Investigator: Project 7; Consulting Investigator: Projects 8, 10; Co-investigator: Project 13 Consulting Investigator: Project 5 Consulting Investigator: Project 3 Investigator: Project 6 Data Safety Monitor Project 9 Collaborator: Project 6 Co-Investigator: Project 1 1 Collaborator: Project 6 Co-Director;Project 1 (U54); IRC Steering Committee; Consulting Investigator: Project 1 (U54) Co-Investigator: Project 1 3 (P30); Investigator: Project 7; Consulting Investigator: Projects 8, 9 Co-Chair, Bioethics Committee, Consulting Investigator: Project 1 (U54) PI: Project 4; Consulting Investigator: Projects 9, 11 Continuation Format Page Levine, Robert MD RJLEVINE Maciejewski, Paul PhD pMaciejewski Mayes, Linda MD LMAYES Mazure, Carolyn PhD CMAZURE McKee, Sherry SMCKEE Miller, Perry PhD MD PMILLER Nairn, Angus PhD ANAIRN Neumeister, Alex PhD ANEUMEISTER Nolen-Hoeksema, Susan SNOLEN PhD Olausson, Peter PhD JPOLAUSSON O'Malley, Stephanie OMALLEYS Picciotto, Marina PhD marina Piomelli, Daniele PhD dpiomelli PharmD Potenza, Marc MD PhD marcnpotenza Rajeevan, Nallakkandi NRAJEEVAN PhD Rosenbaum, Sara JD Rounsaville, Bruce MD . BROUNSAVILLE Ryan, Sheryl MD SHERYLRYAN Santos, Laurie PhD LSANTOS Sherwin, Robert MD rsherwin PHS 398/2590 (Rev. 09/04, Reissued 4/2006) Yale University Yale University Yale University Yale University Yale University Yale University Yale University Yale University' Yale University Yale University Yale University Yale University University of California, Irvine Yale University Yale University George Washington University Yale University Yale University Yale University Yale University Page 6 Chair Bioethics Steering Committee; Data Safety Monitor;Consulting Investigator: Project 1 (U54) Co-Pi: Project 1 2 (R25) Co-Pi: Project 6;IRC Steering Committee PI: Project 12 (R25);Co-Chair IRC Steering Committee;Co-Director: Project 1 (U54) PI: Project 9; Consulting Investigator: Projects 2,4,5,11; Investigator: Project 7 Consulting Investigator: Project 13 (P30) Consulting Investigator: Project 14 PI:Project 8;Consulting Investigator: Project 4; Collaborator: Project 6;Investigator: Project 7 Investigator: Project 6 Co-l: Project 3 Co-l: Project 9;Consulting investigator: Projects 1 (U54), 3, 7, 10, 11; IRC Steering Committee Consulting Investigator: Project 9 PI: Project 5; Consulting Investigator: Project 2, 14 (P30) PI: Project 7; Consulting Investigator: Project 3, 4, 6, 8, 9 Co-l: Project 13 (P30) Policy Consultant Co-Director: Project 1 (U54);IRC Steering Committee;Consulting Investigator: 2, 10 Consulting Investigator: Project 6 Consulting Investigator: Project 4 IRC Steering Committee, Consulting Investigator: Project 1 (U54) Continuation Format Page JSINDELAR Yale University DANASMALL Yale University MSOFUOGLU Yale University Sindelar, Jody PhD Small, Dana PhD Sofuoglu, Mehmet MD PhD Soufer, Robert MD Talley, Sandra PhD RSOUFER Yale University STALLEY Yale University Taylor, Jane PhD JANE TAYLOR Tebes, Jacob PhD JTebes Tice, Diane PhD DIANNEJTICE Vroom, Victor PhD WROOM Wang, Fei PhD FWANG Yeckel, Mark PhD MYECKEL Zhang, Heping PhD Heping Zhang Yale University Yale University Florida State University Yale University Yale University Yale University Yale University Zonana, Howard PhD Yale University Co-Director: Project 1 (U54);PI: Project 11; Co-Investigator: Project 1 3 (P30); Consulting Investigator: Projects 12 (R25) Investigator: Project 7 Co-Investigator: Project 9 IRC Steering Committee; Consulting Investigator: Project 1 (U54) Consulting Investigator: Project 1 (U54), Project 13 (P30) PI: Project 3;Consulting Investigator: Projects 2, 4, 5, 6 Project Leader of IRCSSA Evaluation; Co-l: Project 1 (U54) Co-Pi: Project 10 IRC Team Consultant: Project 12 (R25) Post-Doctoral Associate: Project 13 (P30) Co-Pi: Project 2 Co-Pi: Project 1 3 (P30) Data Safety Monitor: Project 1 (U54), Project 1 3 (P30) PHS 398/2590 (Rev.09/04, Reissued4/2006) Page 7 Continuation Format Page 1:U54 Interdisciplinary Research on Stress, Self-Control &Addiction Principal Investigator/Program Director (Last, First, Middle): SINHA, Rajita RESEARCH GRANT OVERALL TABLE OF CONTENTS Page Numbers Project 1 of 14: Interdisciplinary Research on Stress, Self-Control &Addiction (Face Page) 1 U54 Description, | {
"pile_set_name": "NIH ExPorter"
} |
Vaccination is a potent and cost-effective countermeasure to the threat of bioterrorism agents, and there is intense interest in the design and use of adjuvants to enhance immune responses to vaccines. This is particularly important for vaccines that are based on subunits from bioterrorism agents, since these antigens are usually not highly immunogenic by themselves. Recent work has shown that the C3d component of the cellular complement pathway can function as an adjuvant to selectively enhance antibody responses to a foreign antigen, as well as accelerated affinity maturation of antibodies. Our central hypothesis is that viral vectors can be engineered to be more potent by expressing antigen covalently linked to C3d. Enhanced potency would allow viral vectors to be used at lower and safer doses to obtain better responses than that seen at higher doses of conventional viral vectors. In addition, this approach would allow viral vectors to be attenuated to a higher degree and still maintain potency. We will test this hypothesis by determining if C3d functions as a molecular adjuvant when expressed from vectors based on the noncytopathic paramyxovirus Simian Virus 5 (SV5). Our recent published work has shown that rSV5 has inherent properties that could be exploited to create novel vaccine vectors. In Aim 1, we will test the hypothesis that C3d can function as a selective adjuvant for enhancing respiratory tract antibody responses to Y. pestis F1 antigen when delivered by an rSV5 vector, rSV5 vectors expressing either Y. pestis F1 antigen alone or F1 linked to C3d will be tested in our well-developed mouse model system for viral growth properties, the degree of respiratory tract inflammation, and for cytokine expression and cellular infiltration. Timecourse assays and dose-response experiments will test the potency of rSV5 vectors that express F1 or F1-C3d by determining the antibody response and T helper response following respiratory tract infection. In Aim 2, mice will be infected with various combinations of soluble F1 or F1-C3d along with rSV5-F1 or rSV5-F1-C3d to establish the stage of respiratory tract prime-boost protocols that is best served by C3d: prime only, boost only or both stages. In Aim 3, we will compare the relative adjuvant effect of C3d when delivered by virus vector or by DNA vaccination. At the conclusion of these studies, we will have developed a powerful new class of viral vectors to elicit a more rapid and potent antigen-specific antibody response in the respiratory tract. | {
"pile_set_name": "NIH ExPorter"
} |
DESCRIPTION: a. Candidate: Dr. Karen M. Frank received her M.D. and Ph.D. degrees from the University of Pennsylvania in 1994. She has been a clinical pathology resident and research fellow at Brigham and Women's Hospital and Children's Hospital since 1994. She served as Chief Resident in clinical pathology at Brigham and Women's Hospital from January 1996 to June 1996. Dr. Frank listed six published papers, in which she was first author on four. The candidate's goal is to perform immunology research at a major medical center with an academic faculty appointment, and with a joint appointment in a tertiary care hospital. b. Mentor: Dr. Frederick Alt is the Charles Janeway Professor of Pediatrics and Professor of Genetics and Pediatrics at the Harvard Medical School, and an Investigator of the Howard Hughes Medical Institute at the Children's Hospital. c. Career Development Plan: The candidate will devote 85-90 percent time to research. She will learn new techniques in molecular biology, increase her knowledge of immunology, and develop skills in experimental design and presentation of research results. A modest amount of time will be devoted to teaching and to learning to manage a laboratory. d. Research Plan: Dr. Frank proposes to investigate the function of the newly cloned gene XRCC4 in the V(D)J recombination process. XRCC4 was isolated in the mentor's laboratory on the basis of its potential involvement in V(D)J recombination and double stranded (DS) DNA break repair. The hypothesis of the project is that the XRCC4 protein plays a direct role in the V(D)J recombination process. Three approaches will be used to address the hypothesis. In Specific Aim 1, Dr. Frank will attempt to determine functional domains of XRCC4 by mutational analysis and by examining the evolutionary conservation of amino acid sequence. Dr. Frank will also try to determine if XRCC4 is phosphorylated and whether phosphorylation has any role in its function. In Specific Aim 2, Dr. Frank proposes to investigate whether XRCC4 associates with known V(D)J recombination components such as RAG-1, RAG-2, Ku 80, Ku 70 and DNA-PKCS and other newly discovered proteins. In Specific Aim 3, Dr. Frank will generate XRCC4 knockout (KO) mice and study the role of XRCC4 in lymphocyte development and in DNA repair. e. Environment and Institutional Support: The training will take place in Dr. Alt's laboratory in the Enders Research Building of the Children's Hospital. | {
"pile_set_name": "NIH ExPorter"
} |
There is no definitive evidence regarding how precursor cells in the neuroepithelium switch from generating neuroblasts to generating glioblasts. Studies on the generation of glia cell types, in particular astrocytes, have been hindered by the lack of markers for their precursors and by their extensive migratory pathways. The principal investigator speculates that the cell-type switched to glioblast is distinguished by the differential expression of specific adhesion and extracellular matrix (ECM) molecules, reflecting the differential expression of transcription factors. The developmentally controlled expression of the aggrecan gene in brain ventricular zones during the active period of neuronal migration and gliogenesis suggests an important role for this molecule in neuronal-glial interactions. However, the specific functions and interactions in which aggrecan is involved in vivo remain unclear and will require further investigation. On the basis of the investigator's preliminary results, two hypotheses can be put forth with respect to the function of aggrecan during this process: 1) changes in the composition and/or structure of the ECM may influence the switching of precursor cells differentiating from neuroblast to glioblast cell-fates, or may influence the final glial phenotype (oligodendrocyte or astrocyte) that the precursors can become, and 2) alternatively, aggrecan may regulate the level of cell proliferation or affect the migration of the precursor cells which populate the brain. In order to distinguish between these possibilities a detailed study of aggrecan expression relative to that of different cell type markers, as well as of modifications in cell fate or proliferation associated with the lack of aggrecan expression in the aggrecan-deficient nanomelic mutant, will be the main focus of this proposal. | {
"pile_set_name": "NIH ExPorter"
} |
The goal of this project is to determine the molecular mechanisms which isolate pathways of transcriptional regulation, using the unicellular eukaryote Saccharomyces cerevisiae. We have identified two pathways of transcriptional regulation which show many parallels. The HO and CTS1 genes show identical patterns of cell cycle regulation, with expression in late G1 phase of the cell cycle. These genes are regulated by zinc-finger containing transcription factors, SWI5 and ACE2. These two transcription factors show identical patterns of cell cycle regulated transcription and cell cycle regulated nuclear localization. Furthermore, the DNA binding domains of SWI5 and ACE2 are identical, and they both bind to the HO promoter in vitro. However, despite these many similarities, SWI5 and ACE2 regulate different genes in vivo. SWI5 activates transcription of HO but not of CTS1, and ACE2 activates CTS1 but not HO. We propose to investigate the mechanisms which isolate the SWI5/HO and ACE2/CTS1 pathways. We have data supporting two distinct models for regulation, and these two models are not mutually exclusive. The first model proposes that different negative regulators bind to the HO and CTS1 promoters, and that SWI5 and ACE2 differ in their ability to overcome the repressive effects of these promoter specific negative regulators. In vivo analysis of promoter constructs provides support for this model. The second model proposes that additional factors are required for specificity in promoter recognition, and that cooperative interactions between factors contributes to promoter binding and pathway isolation. In support of this model we have identified a factor which binds cooperatively, along with SWI5, to the HO promoter. We postulate the existence of an analogous factor which is needed for specific promoter recognition of the CTS1 promoter by ACE2. Experiments are proposed to analyze the interactions of these factors with sites on DNA and with each other, and to test models of regulatory control. The factor that binds DNA cooperatively with SWI5 has been purified. The gene encoding this factor has been cloned, and we know this factor is a homeodomain protein. Cooperative interactions have not been observed before between a zinc finger protein and a homeodomain protein, and we propose to characterize their interaction. Experiments are proposed to identify regions of these proteins required for cooperative interactions. | {
"pile_set_name": "NIH ExPorter"
} |
Although it is well-established that schedule-induced drinking occurs on intermittent schedules of food reinforcement, the mechanism by which drinking arises is still a mystery. The proposed experiments will test four major hypotheses. First drinking is a conditioned response that is controlled by the same basic laws as other conditioned responses. Second, drinking arises in anticipation of upcoming food delivery rather than occurring as a reaction to the previous pellet. Third, competition occurs between drinking and the operant, which can result in the displacement of drinking to the early portion of the interpellent interval. Finally, drinking may be controlled by timing mechanisms in much the same way as operant responses. The experimental results will be interpreted with three major theories of timing to determine whether the temporal locus and duration of bouts of drinking may be theoretically modeled in the same manner as operant responses such as lever pressing. If necessary, modifications in the theories will be made to accommodate the results of the proposed experiments. | {
"pile_set_name": "NIH ExPorter"
} |
Various beta adrenergic agonists that increase the accumulation of cAMP are known to induce pineal N-acetyltransferase (NACT) activity. Even though beta adrenergic receptors control both cAMP and cGMP accumulation, cGMP has no apparent role in the increase of NACT. Histochemical localization of cyclic nucleotide indicates that even though most of the cGMP is localized in the nerve innervating the gland it also appears to be present in pinealocytes, even in denervated glands, and catecholamines are markedly decreased. It is possible that cGMP may play a role in the catecholamine and serotonin secretion from the nerve ending in the pineal gland. Studies in isolated pinealocytes from newborn rats indicate that only cAMP is increased 100-200 fold by various catecholamines which is associated with the induction of NACT. cAMP appears to be necessary for maintaining the activity of NACT since lowering of cAMP causes an immediate lowering of the enzyme activity in the pinealocyte and is reversed by dibutyryl cAMP. | {
"pile_set_name": "NIH ExPorter"
} |
DESCRIPTION: The proposed work attempts to define the pathway mediating neuromodulatory effects of natriuretic peptides which produce a variety of hypotensive effects including diuresis, vasodilation, and inhibition of both steroidogenesis and adrenergic neurotransmissions. Past work found natriuretic peptides to inhibit neurotransmission via "clearance" (ANP C) receptors coupling to a pertussis toxin sensitive process (that does not involve a reduction in cytoplasmic calcium concentrations. It is induced catecholamine release. The hypothesized pathway includes (ANP C) receptors interacting with GTP binding proteins to suppress phospholipase C activity, protein kinase C activity and catecholamine release. Experiments will be performed in a rat pheochromocytoma adrenergic cell line, PC12 cells. The PC12 cells allow monitoring of the complete sequence of proposed events (i.e., receptors, GTP binding proteins, phospholipase C activity, protein kinase C activity, and catecholamine release). The specific aims include: 1) assessing the contribution of [ANP C receptors to neuromodulatory effects of natriuretic peptides; 2) assessing the contribution of GTP binding proteins to neuromodulatory actions of natriuretic peptides; and 3) assessing the involvement of phospholipase C and protein kinase C in neuromodulatory DA release actions of natriuretic peptides. The first specific aim will examine effects of both ANP C receptor activation and ablation on subsequent events in the proposed pathway. The second specific aim tests for coupling of GTP binding proteins to the ANP C receptor, phospholipase C and subsequent events in the hypothesized pathway. The third specific aim tests whether suppressed phospholipase C and protein kinase C activities are essential for neuromodulatory influences of natriuretic peptides.] These results will impact on the natriuretic peptide field by identifying alternative signal transduction pathways to guanylyl cyclase activation, the pathway currently perceived to mediate all natriuretic peptide actions. The primary contribution will involve a critical assessment of the natriuretic peptide receptor involved in mediating neuromodulatory effects and the coupling of this receptor to GTP binding proteins. The proposed work will test if the (ANP C) receptor specifically mediates neuromodulatory actions of natriuretic peptides. | {
"pile_set_name": "NIH ExPorter"
} |
Five years after its formation, the Clinical and Translational Science Award (CTSA) at the Univeristy of North Carolina at Chapel Hill (UNC), the North Carolina Translational and Clinical Sciences Institute (NC TraCS), has become the integrated, campus-wide home for clinical and translational research at UNC. The overall goal of this U54 Cooperative Agreement application is to combine the research strengths, resources and opportunities at UNC and new partner, RTI International (RTI), to build on the foundation established in the CTSA's last five years. NC TraCS .will work to improve human health by accelerating clinical and translational research from health science discovery to dissemination to patients and communities. NC TraCS will strive to overcome the well-documented barriers to the effective, sustained translation of research discovery along the translational research continuum by improving efficiency, training a research workforce and exporting successful, validated methods developed in NC TraCS to other CTSA institutions and the public. The proposed CTSA will specifically (1) amplify a thriving CTSA program to the point that it supports the full spectrum of clinical and translational research; (2) leverage CTSA resources and institutional strengths to create a critical, sustained focus on three strategic initiatives: (a) next-generation technologies to transform the very nature of clinical research and practice, (b) new paradigms and resources to accelerate drug development, and (c) robust comparative effectiveness research studies to provide definitive evidence of the benefits and harms of tests and treatments; and (3) train, support and incentivize the next generation of clinical and translational science researchers. These goals will be accomplished by three resources, six services, integrated and streamlined from the original 11, and the three strategic initiatives. UNC and RTI now have a unique set of research and training resources to support the full range of clinical and translational research, from basic science to clinical application to policy change. NC TraCS will leverage the opportunity of this CTSA application to garner >$60 million dollars in matching institutional support to extend our capabilities to meet the three aims. In partnership with RTI, NC TraCS will take advantage of the resources it has created and nourished during the past five years to quickly and effectively bring the fruits of research to patients across the state, as well as nationally through the CTSA Consortium. | {
"pile_set_name": "NIH ExPorter"
} |
Recent years have witnessed a significant increase in the use of coronary revascularization (CABG or PCI). The total number of CABG and PCI procedures performed in the US has increased approximately 2 fold between 1990 and 2001 to more than a 1.6 million per year (American Heart Association, 2004 update) with $33 billion in associated health care expenditures (HCUP, 2004). 1 less studied aspect of delivering these procedures is cost and its relationship to outcomes of cardiac care. The health care system is again under scrutiny for the significant increases in health care expenditures and quality has become at the forefront of policy makers' and the public's agenda. As such, it becomes important to explore the determinants of cost and whether an association exists between costs and outcomes of care, especially with the increased interest in pay-for-performance approaches. This study aims to address this issue through examining 2 'popular' procedures, CABG and PCI. More specifically, the study has the following aims: (1) Examine the association between risk-adjusted hospital-level costs and outcomes of coronary revascularization procedures. (2) Investigate the effect of patient, hospital and market characteristics on hospital-level costs of coronary revascularization procedures. The significance of this study is in its attempt to explore whether a pay-for-performance opportunity exists for coronary revascularization procedures. Through measuring the association between costs and outcomes, 1 of 2 scenarios may be revealed through the study results: 1) no association between costs and outcomes or 2) an association between costs and outcomes. If the first scenario was proven to be true, that may have a significant impact on the health care reimbursement mechanisms. Public and private payers may choose to selectively contract with low-cost hospitals without fear of adverse outcomes. On the other hand, the second scenario may allow private and public payers to employ a pay-for-performance scheme through which high-quality hospitals are reimbursed at higher rates to subsidize the additional costs or provide incentives to maintain high quality. Another way to examine the findings is through grouping hospitals in 1 of 4 categories: 1) low-cost low-quality, 2) low-cost high quality, 3) high-cost high-quality and 4) high-cost low-quality. Hospitals that will be in the second and third category may be eligible for additional incentives (reimbursement) under a pay-for-performance scheme. [unreadable] [unreadable] | {
"pile_set_name": "NIH ExPorter"
} |
This proposed research is descriptive in nature. It is designed to characterize the effects of a sucrose diet, obesity, and pregnancy on miniature swine which have been selectively bred for reduced glucose tolerance. The time course of developing insulin resistance and further changes in glucose tolerance and insulin response will followed in vivo, and in vitro in isolated adipocytes. Lipid profile changes will be monitored. Capillary basement membrane thickness will be periodically assessed as well as the rate of tissue healing and vascularization of implanted plastic sponges. In the final year 1/2 of the animals will be placed on a weight reduction regime using the same diets to establish the rate at which normal insulin resistance returns. At the conclusion of the study the animals will be necropsied and examined for signs of cardiovascular lesions as well as pancreatic morphologic changes. | {
"pile_set_name": "NIH ExPorter"
} |
A reversible, H+-transporting ATPase catalyzes ATP synthesis during oxidative phosphorylation. The enzyme is made up of two sectors. F1 lies at the membrane surface, and on isolation catalyzes ATP hydrolysis. F-o extends through the membrane and functions in H+ transport. When the two sectors are properly coupled, ATP synthesis/hydrolysis is coupled to H+- translocation. The F1F-o complexes of mitochondria and Escherichia coli are very similar so general features of one system should be applicable to the other. In E. coli, F1 is composed of 3 pairs of alpha-beta subunits which form catalytic sites, and a single copy of the gamma, delta and epsilon subunits which link F1 to F-o. F-o is composed of 3 subunit types in an a1b2c10 ratio. Subunit c is thought to play a direct role in H+ transport, and in coupling H+ transport to ATP synthesis. It is a protein of 79 residues, folding in the membrane as a hairpin of two alpha-helices. Asp61 (centered in transmembrane helix-2) is thought to mediate H+ transport; H+ transport is thought to alter the loop region of subunit c and initiate a conformational change which ultimately promotes ATP release from alpha-beta subunits in Fl. We have shown that the essential carboxyl of subunit c can be moved from helix-2 to helix-1 and function retained. Analysis of suppressor mutants, optimizing function in the carboxyl-transposition mutant, led to identification of a transmembrane helical surface in subunit a that we now postulate interacts with subunit c. Secondly, polar loop mutants which uncouple H+-transport from ATP synthesis in F1 were characterized. Second site revertants to one of the uncoupled mutants map to a single residue of subunit epsilon. Here, we will better define, by both genetic and physical methods, the interaction between subunits c and a during H+ translocation, and the coupling interaction between the loop of subunit c and subunit epsilon. Additional aims include determination of the solvent accessibility and pKa of Asp6l in situ, and the relationship of this pKa to H+ binding during transport. We have shown that subunit c folds in a chloroform-methanol-H2O solvent much like it is predicted to fold in situ. Further, the unique chemical reactivity of Asp6l is retained. We have derived a low resolution model for the folding of subunit c in this solvent using 2D NMR methods and a novel approach using paramagnetic broadening with a nitroxide derivatived Asp6l. We propose to complete a high resolution solution structure by heteronuclear 3D and 4D methods and to test aspects of the solution model by mutagenesis. This structure will be compared to the structure in detergent micelles, and attempts made to study structural features of c-c and a-c aggregates in an organic solvent mixture. An understanding of structure-function relationships in membrane proteins is fundamental to many problems in biology and medicine. Few intrinsic proteins are understood in any detail. The H+ ATP synthase is central to all cellular functions, i.e. it makes the ATP. Abnormalities in the function of this enzyme, or other mitochondrial respiratory enzymes, result in human disease. | {
"pile_set_name": "NIH ExPorter"
} |
Studies on endogenous sequences of FeLV and RD114: Determine the sites of integration of FeLV and RD114 related sequences by transferring individual cat chromosomes to a mouse background; and testing for the presence of the proviral sequences in situ and other molecular hybridization tests. Map the provirus in relation to other markers in the cat genome. Studies on exogenous FeLV: Conduct somatic cell hybrids between mouse and exogenously infected cat cells, and test the synteny of the exogenous FeLV sequences with specific chromosomes and marker genes. Prepare hybrids of mouse x Asian cat (Felis viverrina) tissue culture cells infected with FeLV+ cat chromosomes free of endogenous FeLV sequences. Test for integration site of exogenous FeLV. | {
"pile_set_name": "NIH ExPorter"
} |
Nitric oxide (NO) participates in the physiology and pathophysiology of every organ system. Of most importance, NO acts as an major mechanism of vasodilation, both under physiologic and toxic conditions. During the first 2.5 years of this project, we have identified several steps in the induction process of iNOS (microtubules, PTK, PKC, NF-kappaB), as well as mechanisms of NO-mediated control of soluble guanylate cyclase, the enzyme responsible for mediating most of the actions of NO. Recently, we also cloned 5.0 Kbases of the rat iNOS promoter and sequenced the 3.2Kbases apparently responsible for transcriptional control of the gene. In the proposed studies, we will investigate the molecular mechanisms involved in the promoter- dependent up-regulation of iNOS, and down-regulation of cNOS in vascular cells during immunological challenges. We hypothesize that NF-kB binding sites in the iNOS promoter are major elements in iNOS up- regulation in responding to immunomodulators; however, other factors (e.g., CRE) are also involved in iNOS expression; we further hypothesize that a similar NF-kappaB dependent signal transduction pathway is responsible for immunomodulator-induced endothelial cNOS down-regulation. Specifically, we will investigate 1) the essential element s of rat iNOS promoter in responding to cytokines and LPS stimulation, 2) the mechanism by which cAMP synergistically up-regulates the iNOS expression in rat ASMC in responding to cAMP and LPS, 3) the mechanism by which cytoskeleton disrupting agents decrease the iNOS expression and 4) differences between the rat and human iNOS promoter responsible for the weak iNOS up regulation in response to LPS and cytokines in human cells. Furthermore, 5) we will quantify LPS-, IL-1-, and TNF-induced oxidant stress and thiol depletion and determine the contribution of oxidants to reduced NO generation in LPS, IL-1 and TNF-treated EC, 6) we will investigate the kinetics and mechanisms of LPS, IL-1 and TNF-induced activation of protein kinases, NF- kappaB, cytoskeletal reorganization and determine the contribution of these mechanisms to cNOS down-regulation in cultured EC, 7) we will investigate the mechanisms of iNOS up-regulation in LPS- induced in rats, in vivo. Our findings should help clarify the role and mechanisms of iNOS activation and cNOS down-regulation under toxic conditions. | {
"pile_set_name": "NIH ExPorter"
} |
PROJECT ABSTRACT Nucleotides and nucleotide sugars are released in regulated fashion from both excitable and non- excitable cells and act as extracellular signaling molecules to regulate a remarkably broad array of physiological responses ranging from neurotransmission to epithelial ion transport to platelet aggregation. At least fifteen different P2 receptors recognize extracellular nucleotides as their cognate agonists, and eight of these comprise the family of metabotropic G protein-coupled P2Y receptors. Although signals emanating from P2Y receptors convey important physiological and pathophysiological responses in essentially all tissues, physiological and molecular understanding of the action of these receptors has lagged, largely due to lack of reliable molecules to probe unambiguously the actions of single receptor subtypes. An antagonist (clopidogrel; Plavix) of the platelet ADP-activated P2Y12 receptor has made an enormous impact in modern therapeutics, and the potential for drugs that clinically target other G protein-coupled P2Y receptors is both very large and as yet unrealized. Our research program has developed and applied the first selective and high affinity agonists, antagonists, and radioligands for the study of P2Y1 receptors. Several of these molecules are now commercially available and are key reagents used by many laboratories to elucidate mechanism in this field of research. The long-term goal of our research is to identify subtype-selective agonists and antagonists for each of the nucleotide-activated P2Y receptors and to apply these molecules to increase molecular, physiological, and pathophysiological understanding of P2Y-receptor-dependent signaling. We recently have made considerable progress in defining structure activity relationships at the uridine nucleotide- activated P2Y2, P2Y4, and P2Y6 receptors and at the nucleotide sugar-activated P2Y14 receptor. Included in this progress is promising new insight for development of selective, high affinity antagonists of these receptors. Our proposed research plan will expand on these discoveries in important new directions. In the first specific aim we will synthesize and characterize selective high affinity agonists and antagonists of the UTP-activated P2Y2 and P2Y4 receptors. In the second specific aim we will synthesize and characterize selective high affinity agonists and antagonists of the nucleotide sugar- activated P2Y14 receptor and agonists of the UDP-activated P2Y6 receptor. Completion of the research described here will provide much-needed molecular tools to selectively activate and block uridine nucleotide- and nucleotide sugar-activated P2Y receptors with high affinity and selectivity. | {
"pile_set_name": "NIH ExPorter"
} |
Pathologic hyperprolactinemia in men can be associated with serious reproductive disorders including impotence and hypogonadism. We have developed an animal model for studying the effects of hyperprolactinemia on male reproductive functions. Transplantation of four pituitaries under the kidney capsules of adult male rats or mice produces chronic elevation of peripheral PRL to supraphysiological levels, drastic deficits of copulatory behavior and a reduced ability to release gonadotropins. We will utilize this experimental model to define the mechanisms by which hyperprolactinemia affects reproductive functions in the male. By studying mating behavior in males given pituitary grafts and a drug which inhibits prolactin (PRL) release, we will determine whether deficits in copulatory behavior can indeed be explained by elevated PRL levels. We will also determine whether the stimulation of adrenal growth and function in pituitary grafted males accounts for or contributes to the suppression of mating behavior. Furthermore, we will determine whether replacement therapy with LHRH, hCG or testosterone can stimulate copulatory behavior in hyperprolactinemic males. In a separate series of experiments, we will define the effects of hyperprolactinemia on endogenous LHRH levels, on hypothalamic sensitivity to electrical stimulation and on pituitary responsiveness to LHRH. We will also measure the levels of gonadotropins and gonadotropin receptors, as well as the responsiveness of the testis to gonadotropins in pituitary grafted males. During the proposed period of support, we will initiate pilot studies of hypothalamic, pituitary and testicular function in men with hyperprolactinemia. | {
"pile_set_name": "NIH ExPorter"
} |
Micro array analysis of genome expression data is an emerging technology that allows biologists to measure patterns of gene expression across a large portion of a single genome in one measurement. The technology is currently in the process of commercialization, and it is expected that organizations that adopt the technology will accumulate large databases of results of micro array analyses. The very high dimensionality of micro array data means that existing data base indexing schemes are not well suited to the problem of providing efficient content based searches of large data bases of microarray data. The work proposed here will develop a new database indexing scheme that is specifically designed for efficient searches of microarray databases. The innovative feature of the design is the combination of a feature map, which maps the very high dimensional data space onto an feature space of lower dimensionality, and an index on the lower dimensional feature space that uses a binary space partition tree to organize the geometry of the feature space. AIMS has successfully applied a similar methodology to indexing problems involving data bases containing high dimensional signal data from radar pulses. The software that was developed for these applications would form the basis for the software that would be implement the micro array index. To investigate the performance of the proposed indexing scheme a model using metabolic networks will be developed which will be designed so as to produce synthetic micro array data with statistical characteristics similar to those of actual experimental data. PROPOSED COMMERCIAL APPLICATIONS: This research will provide a fundamental database component that will increase the usefulness of large databases of micro array assays of genome expression in tissue. No good alternative technologies currently exist. Any company wishing to develop a system that will efficiently query the information content in large microarray databases will require an indexing scheme such as the one proposed here. | {
"pile_set_name": "NIH ExPorter"
} |
Program Summary Stroke is the leading cause of adult disability. As the population ages, the incidence of stroke is increasing. Improvements in acute stroke care mean that the death rate from stroke is declining. Stroke has thus changed into a disease of chronically disabled survivors. There is no medical therapy that promotes recovery in stroke. This proposal develops a stem cell therapy for a common subtype of stroke, subcortical or ?white matter? stroke. White matter stroke occurs in the regions of the brain that carry connections, is the most age-associated in its incidence, and is the second leading cause of dementia (termed vascular dementia). The proposed studies characterize the molecular and cellular process in tissue integration and wound healing of an hiPSC-derived cell in brain repair after white matter stroke. This cell is differentiated from an induced human pluripotent cell (hiPSC cell) into an immature glial cell, and is termed an hiPSC-glia enriched progenitor (hiPSC-GEP). Preliminary studies indicate that transplantation of hiPSC-GEPs into a mouse model of white matter stroke enhances behavioral recovery compared to other hiPSC types, and that this behavioral recovery effect is associated with improvement in the MRI appearance of white matter stroke. The studies in this grant will fully determine the phenotype and tissue integration of hiPS-GEPs, the transcriptional profile of hiPSC-GEPs and the cells of the surrounding stroke environment as these two populations re-organize and inter-relate over time after stroke, the effect of hiPSC-GEPs on stroke using in vivo MRI to track tissue repair, and the efficacy of hiPSC-GEPs in translationally relevant stroke conditions, such as in the aging brain. These studies use an innovative platform that includes a new mouse model, in vivo MRI, Drop-seq single cell transcriptional profiling, and viral reporter and gain and loss of function approaches. The proposed studies bring together a neuroscientist/neurologist and stem cell biologist to develop a novel therapy in a disease with increasing prevalence and no current therapy. | {
"pile_set_name": "NIH ExPorter"
} |
The Maternal Fetal Medicine Network is one of the premire groups of clinical investigators in the country. The Columbia Center has been privileged to be a member of this research collaborative for over 15 years and is now submitting a competitive renewal application for the next 5 years. The Columbia Center is composed of 6 large perinatal centers working together under a single administrative structure administered by Columbia University. The centers have a long and successful history of collaborative multicentered research. During the last 5 year period of the MFMU, the Columbia Center has ranked first in recruitment. Columbia Center faculty have designed and led major trials, performed ancillary studies and secondary analysis, served on subcommittees, and successfully presented concepts. The Columbia Center provides over 25,000 deliveries per year of which over 30% are high risk; has 33 MFM physicians, several of which have the protected time and expertise to be involved in Network activities; tertiary level NICUs with neonatologists experienced in Network studies; well established infant follow-up programs; and over 40 nurses, recruiters and staff dedicated to network activities. | {
"pile_set_name": "NIH ExPorter"
} |
We have generated a mouse line with a mutation frequently found in human MYH9 related disease, E1841K. In addition to modeling the defects seen in humans with MYH9-related disease, male, but not female, mice homozygous for the mutation are sterile. The male mice demonstrate severe defects in sperm development. Histological analyses of homozygous testis reveal that spermatocytes arrest at the elongated spermatid stage and aberrant spermatids slough off into the lumen. Some seminiferous tubules contain vacuoles, have a large lumen and often lack germ cells, while others appear normal but completely fail to develop a lumen. Electron micrographs uncover an abnormal flattening and extension of the acrosomal vesicle, which appears to lead to the abnormal elongation of spermatids. TUNEL assays of homozygous testis show an increase in apoptotic germ cells when compared to wild type. Caudal epididymis sections have only a few malformed spermatozoa that are rarely motile. Heterozygous mice show similar, albeit less severe, defects and are fertile. Sperm counts obtained from the caudal epididymis confirm a pronounced decrease in mature sperm in homozygous mice, while the heterozygous mice show a modest decrease when compared to wild type. Collectively, these observations suggest a previously unknown, critical role for NMII-A in spermatogenesis and male fertility. | {
"pile_set_name": "NIH ExPorter"
} |
The applicants propose to study the effects of treatment with a nondepleting CD4 mAb (RIB-5/2) or rapamycin (RPM) + synthetic fibronectin (FN) peptides in sensitized rat recipients of cardiac allografts (Tx). RIB-5.2 mAb or RPM monotherapy abrogates rejection at 24 h and prolongs cardiac Tx survival to >60 days and ca. 50 days respectively. The approach to preventing Tx rejection with FN peptides is based upon observations that : (i) rejection triggers increased intra-Tx deposition of FN, (ii) adhesive FN- MNC associations are decisive for intra-Tx MNC recruitment, (iii) FN-MNC interactions are vital for cell activation/migration/adhesion, (iv) treatment with FN peptides prevents/reverses acute or chronic inflammatory conditions (e.g. rat model of erosive arthritis, TGFB1 knockout mice). The specific aims are as follows: 1. To investigate intraTx expression /function of FNs, and FN-MNC interactions: Cardiac transplants from untreated and RIB-5.2 mAb or RPM treated hosts will be analyzed for FN expression at the mRNA (Northern blotting/RNase protection assay) and protein (immunohistochemistry) levels. The expression of total FN, and FN splicing variants (EIIIA, EIIIB, CS-1) will be correlated with FN receptors on infiltrating MNC. The FN producers will be identified by in situ hybridization. MNC positioning in relation to FNs will be studied by laser scanning confocal microscopy. The adhesive/migratory/differentiation properties of MNC will be tested in vitro on FN-coated substrata. The ability of anti-integrin mAb, FN peptides interacting with integrins or cell surface proteoglycans, and FN fusion proteins to modulate cell functions will be evaluated. The effects of treatment with FN peptides/fusion proteins on in vivo cell migration will be analyzed. 2. To assess the effects of adjunctive treatment with FN peptides on Tx survival and FN-MNC interactions. Individual or combined FN peptide preparations will be administered into RIB-b/2 mAb or RPM treated hosts bearing long-term (>30 day) cardiac Tx. The effects of treatment with FN peptides upon adhesive/migratory/differentiation properties of intra-Tx infiltrating MNC will be evaluated in vitro and in vivo as outlined in Aim 1. 3. To evaluate the effects of applied therapies upon host cell-mediated immunity: Cardiac Tx will be examined by immunohistology to identify infiltrating MNC by phenotypic/activation markers. The binding patters of naive or host LNL to HEV of naive or recipient lymph nodes and cardiac T will be studied: the specificity will be ascertained in mAb inhibition experiments. in vitro LMC, MLR, flow cytometry and in vivo adoptive transfer, allo-Ag challenge experiments will unravel putative feedbacks between immunomoulatory agents and cellular repertoire in host lymphoid organs. 4. To explore the cytokine networks: Intra-Tx expression of cytokines will be examined by RT-PCR and immunohistochemistry. The studies will focus on Th1 (IL-2, IFN- ) vs Th2 (IL-4, IL-10) cytokines, and IL-12; CTL specific granzyme B: FN enhancing growth factor (TGF-B1); neutrophil (MIP-2, KC) or macrophage (MCP-1) stimulatory chemokines. To clarify the relevance of gene products, Tx hosts will be challenged with rIL-2, rIFN- , rIL-12, or cytokine specific Abs. 5. To assess the effects of applied therapies on host humoral immunity: The utilization patterns and isotype switching of allo-Abs will be studied in the circulation (flow cytometry), and at the Tx site (immunochemistry, radial immunodiffusion). The functional significance of host humoral reactivity will be probed by testing sera samples in CDC, ADCC, and in passive transfer studies. The effects of sera on the alloreactive T cell functions will be evaluated in MLR. | {
"pile_set_name": "NIH ExPorter"
} |
Macromolecular structure, dynamics and polyelectrolyte properties of large biological polymers, in particular, polynucleotides and nucleic acids are being studies by electric-field induced dichroism and birefringence methods. Theoretical and computational methods supplement the experimental work. The current research is a response to the fact that the knowledge of the structural effects of specific base-pair sequences on DNA translation and replication is still at a primitive stage. Only one or two biologically significant protein-DNA complexes from which such structural effects could be inferred have been crystallized and their structure determined. Using electro-optic birefringence and dichroism, it is now possible to quantitatively explore DNA structures in solution, albeit with less resolution than x-ray diffraction of crystals, but uninhibited by the problem of forming crystalline complexes. The two principal projects currently being pursued are the structural effects of the triplet sequence CAC/GTG and the flexibility of the A form of DNA. | {
"pile_set_name": "NIH ExPorter"
} |
The plant Cannabis and its extracts have long been used to treat pain. However, their psychotropic effects[unreadable] have prevented their widespread use in medicine. Their effects have been attributed to activation of[unreadable] cannabinoid receptors, CB1 and CB2. CB1 are highly expressed in the CMS, while CB2 is exclusively[unreadable] expressed on immune cells. Recent studies from our lab have demonstrated for the first time that naturally[unreadable] occurring plant cannabinoid, cannabidiol (CBD), which is non-psychoactive, can induce apoptosis in immune[unreadable] cells and is very effective in the treatment of autoimmune hepatitis (AIM). In the current study, we will also[unreadable] test the central hypothesis that CBD induces apoptosis in T cells through death receptor and/or[unreadable] mitochondria! pathways which are responsible for causing immunosuppression. Our studies will address the[unreadable] mechanism by which CBD may serve as a therapeutic modality in the treatment of autoimmune diseases,[unreadable] specifically staphylococcal enterotoxin B (SEB) and ConA-induced hepatitis (CAM), considered to be[unreadable] experimental models for human AIM. Liver disease is a major cause of morbidity and mortality and also the[unreadable] prognosis is poor. In many liver diseases including viral hepatitis, AIM and alcoholic liver disease, activated T[unreadable] lymphocytes and macrophages appear to play an important role in liver damage. AIM is an inflammatory liver[unreadable] disease that is primarily triggered by T cells. To address the effect of CBD on hepatitis, we will pursue the[unreadable] following Aims: # 1: We will use of CB1, CB2, CB1/CB2 and VR1 knockout (KO) mice to address the role of[unreadable] CB1, CB2 and vanilloid receptors in signaling CBD-induced apoptosis in T lymphocytes. # 2: We will identify[unreadable] the mechanisms of CBD-induced apoptosis in immune cells in vitro and in vivo, specifically the role of death[unreadable] receptor pathway versus the mitochondrial pathway. # 3: Effect of CBD on SEB-specific V beta8+ T cells and[unreadable] SEB-induced AIM will be investigated. # 4: Use of CBD in treatment of Con A-induced hepatitis and liver[unreadable] injury will be tested. We will investigate the effects of CBD on T cells, NK cells and NKT cell functions and[unreadable] their ability to produce inflammatory cytokines. Whether the effect of CBD is mediated through cannabinoid[unreadable] or vanniloid receptors will be tested.Together, the proposed studies will help in identifying the mechanisms[unreadable] through which a plant derived non-psychoactive cannabinoid, CBD can be used to effectively treat[unreadable] autoimmune hepatitis and liver injury.[unreadable] | {
"pile_set_name": "NIH ExPorter"
} |
Overall Component for the University of Pennsylvania Perelman School of Medicine Morris K. Udall Parkinson's Disease Research Center of Excellence: ?Parkinson's Disease and Dementia? Udall Center Director: J.Q. Trojanowski Udall Center Summary/Abstract: The goals of the competing renewal application for the University of Pennsylvania (Penn) Perelman School of Medicine Udall Center are to elucidate mechanisms of progressive neurodegeneration in Parkinson's disease (PD), especially those that underlie cognitive impairments in PD without and with dementia (PDD) and in dementia with Lewy bodies (LBs) or DLB (Projects I/II) that we refer to collectively as LB disorders or LBD. Udall Center investigators hypothesize that LBD leads to neuron dysfunction and death resulting from the differential transmission of pathologic alpha-synuclein (a-syn) strains leading to the formation of Lewy bodies (LBs) and neurites (LNs) and their neurotoxic effects. Understanding LBD progression and its heterogeneity from molecular to patient levels will advance our ability to develop a precision medicine approach to LBD care and management. Thus, Projects III/IV seek to elucidate the conformationally distinct strains underlying PD, PDD and DLB compared to those underlying multiple system atrophy (MSA) characterized by glial cytoplasmic inclusions (GCIs) formed by misfolded a-syn, while Project I seeks to better clinically manage the diversity of phenotypes within LBD that may result from these strain differences, and Project II extends our analysis of phenotypic diversity to the important area of cognitive decline. Consistent with the mission of the Udall Centers described in the most recent RFA, the vision statement of the Penn Udall Center for the renewal period is to elucidate the progression of PD from normal cognition to cognitive impairment, executive dysfunction and dementia in PDD, as well as disease progression in DLB in addition to neurodegeneration mediated by progressive accumulations of pathological a-syn. The landmark discovery of mutations pathogenic for PD in the a-syn gene, the discovery of pathological a-syn as the disease protein that forms LBs/LNs in PD/PDD/DLB as well as GCI in MSA in addition to evidence for the cell-to-cell spread of a-syn strains places a-syn at center stage for understanding mechanisms of LBD/MSA. The Penn Udall Center renewal seeks to address these key issues in four Projects supported by four Cores including: Administrative Core A: Core Leader (CL) - John Q. Trojanowski; Clinical Core B: CL - Daniel Weintraub, Co-Investigators (Co-Is) - Lama Chahine, Nabila Dahodwala, James Morley, Alice Chen-Plotkin; Neuropathology & Genetics Core C: CL - John Q. Trojanowski; Co-Core Leaders - Edward B. Lee, Vivianna Van Deerlin; Data Management, Biostatistics & Bioinformatics Core D: CL - Sharon Xie, Co-I - Li-San Wang. These Cores support the following Projects to achieve the goals of the Udall Center renewal: Project I ?A Framework for Precision Medicine in Parkinson's Disease?: Project Leader (PL) - Alice Chen Plotkin; Co-I - Dan Weintraub; Project II ?Executive Difficulty in Parkinson's Dementia?: PL - Murray Grossman; Co-Is: Rizwan Akhtar, David Irwin, Corey McMillan; Project III ?Mechanisms of Pathological A-syn Transmission?: PL - Virginia M.-Y. Lee; Project IV ?Pathologic A-syn Strains & Diverse Synucleinopathies?: PL - John Q. Trojanowski; Co-Is - Virginia M.-Y. Lee, Kelvin Luk. | {
"pile_set_name": "NIH ExPorter"
} |
As discussed earlier, we reported last year that halothane metabolism led to an unfolded protein response and subsequent liver injury that appeared to be due in part to a loss of important antioxidant proteins as a result of protein catabolism in the absence of protein synthesis. This year we discovered that other important cellular proteins are also lost from the liver by a similar mechanism, including those involved in liver autophagy. Conclusion: The murine model of halothane-induced liver injury continues to reveal novel mechanisms of DILD. Our findings this year suggest that the unfolded protein response induced by halothane can lead to liver injury by inhibiting hepatocellular autophagy, which has been reported to be an active process in protecting hepatocytes from death caused by a variety of etiologies. It is quite possible that liver injury caused by other drugs might also be mediated by similar pathways involving autophagy inhibition. | {
"pile_set_name": "NIH ExPorter"
} |
The broad, long-term objective of this project is to explore the potential role of ethanol infusion in the vein of Marshall (VOM) in the treatment of atrial fibrillation (AF). AF is the most common sustained rhythm disorder in adults, affects more than 2.5 million Americans, and is associated with significant increases in stroke and mortality. Catheter ablation of AF, consists of pulmonary vein antral isolation (PVAI) and is the most powerful therapeutic strategy to treat AF, achieving normal rhythm in ~80% of patients. Important details of the mechanistic basis of PVAI, the optimal catheter technique, and the risk of complications are incomplete or controversial. The VOM is a left atrial vein branch of the coronary sinus that is the embryonic remnant of the left superior vena cava, and contains important sympathetic and parasympathetic nerves that have been implicated in the genesis of AF. Abnormal ectopic beats originating from the VOM have been shown to initiate AF. The VOM lies in a location in the LA that is normally part of the targeted tissue in PVAI and can be difficult to ablate conventionally due to its thickness (lateral ridge). Of note, it is also between the coronary sinus and the left pulmonary veins, an area commonly ablated to avoid left atrial flutter. It can also run close to the point of contact between the left atrium and the esophagus. A new technique to retrogradely cannulate the VOM was developed and validated in dogs and humans. Once cannulated, ethanol infusion achieves rapid tissue ablation without risk of collateral damage. The potential advantages include: ablation of sympathetic and parasympathetic innervation that promotes AF, ablation of VOM triggers for AF, ablation the lateral ridge from its epicardial side, and rapid tissue ablation from a right-sided procedure. We propose to study details of the mechanistic basis of this technique and to establish its role in the treatment of AF in humans. Specific aim #1 is to establish in dogs the electrophysiological effects of VOM ethanol infusion. We hypothesize that VOM ethanol infusion leads to marked direct and indirect electrophysiological changes in the LA that affect its ability to sustain AF. Direct effects would include of a new ethanol- ablated unexcitable LA area. Indirect effects would include abolition of vagal and sympathetic influences carried by the VOM. Experiments will be performed to delineate these effects. Specific aim #2 is to establish the role of VOM ethanol infusion as a useful adjunct to catheter ablation of AF in humans. Feasibility, safety, ablative effects and procedural impact of VOM ethanol infusion will be tested in patients subjected to conventional AF catheter ablation. Once the feasibility and safety are established, we will assess, in a randomized clinical trial, the value of adjunctive VOM ethanol infusion in de novo AF catheter ablation with PVAI, and in cases of recurrent AF after previous PVAI. PUBLIC HEALTH RELEVANCE: Atrial fibrillation is a significant health problem that causes stroke and increases death rates. Catheter ablation works in its treatment but can be improved. We have developed a new technique that is mechanistically sound and may improve catheter-based treatment of AF. | {
"pile_set_name": "NIH ExPorter"
} |
Although poverty in the United States has decreased since 1959, poverty and its related problems continue to pose important threats to American inner cities. One of the poverty-related problems that has defied solution is substance use and abuse among adolescents living there. Many substance abuse programs have been implemented to serve adolescents living in impoverished inner-city areas. However, they have not been specifically designed to meet the needs of inner-city adolescents living in extreme poverty; nor has their effectiveness with this population been evaluated. The special circumstances of inner-city adolescents require a different approach than is typically adopted. Many experience feelings of disempowerment, hopelessness, and despair that may prevent them from actively engaging drug and alcohol services; and those that do receive substance abuse services return to the same environment that helped create their risk behavior in the first place. The only way to counter these negative forces is to couple substance abuse treatment with an intensive long-term effort to change the neighborhoods themselves, working with residents to build a sense of community and a belief that they can improve the quality of life in their neighborhoods. We have incorporated this approach into our Strengthening Neighborhood Investment Program (SNIP), a community-based substance abuse treatment program funded by CSAT; in fact, community development and outreach are tied directly to the treatment services, with outreach workers taking a role in the treatment and the aftercare that adolescents receive. This program has been implemented in five inner-city neighborhoods in Mobile, Alabama. We propose to conduct a comprehensive evaluation of SNIP, which will add to our understanding of adolescent substance abuse and its treatment in several ways. First, the evaluation will examine the effectiveness of evidence-based treatment modalities for underserved inner-city adolescents whose needs and circumstances are different from those adolescents who are typically served by treatment programs. Second, it will examine the value added by an intensive outreach and community- development component. Third, it will examine the implementation of substance abuse programs in inner-city neighborhoods. Fourth, it will explore the impact of this neighborhood-based program on the metropolitan area system of care for adolescent substance users and abusers. | {
"pile_set_name": "NIH ExPorter"
} |
Congenial anomalies of the kidney and urinary tract are common in childhood and may lead to renal failure, infection, and hypertension. In addition, premature infants experience deficits in renal function because of incomplete renal maturation. For these reasons it is important to understand the mechanisms of normal renal development and of congenial anomalies. Mouse models of human disease serve as useful tools to examine these mechanisms. The applicant has begun to study a model that has early ureteric bud branching defects and an unexpected defect in dorsoventral renal axis formation. Late in development the mice have non-obstructive renal pelvic dilation. This proposal will use the mouse model and organ explant cultures to extend our understanding of the mechanisms leading to renal anomalies and the mechanisms that control normal development. The aims will be to 1) characterize the mechanism leading to pelvic dilation in the mutant; 2) determine the mechanism that controls differentiation of the collecting duct cell subtypes; 3) characterize the mechanism of proximo distal nephron axis formation; 4) to evaluate the potential for environmental regulation of hoax-11 and hold-11 that would produce of a more serve renal anomaly. The applicant for the award is a pediatric nephrologist who has had an interest and previous experience in renal development and who plans a long-term career in studying in the mechanisms of renal organogenesis. The award will allow him to broaden the scope of his research. The applicant has had a productive maundered relationship with a well- established investigator and will obtain help from two other investigators in the institution. The institution has a particularly strong track record in developmental biology and continues to support new research activity in this field. The environment will allow the candidate to achieve his career goals. | {
"pile_set_name": "NIH ExPorter"
} |
Surgical repair of intracardiac defects with pulmonary hypertension is still associated with a 20 to 30 percent mortality, often secondary to right ventricular dilatation and failure. Unloading of the right ventricle may be accomplished by venous unloading only at the expense of left ventricular output. Therefore we are evaluating a unidirectional intra-atrial flap valve that allows right-to-left shunts when right ventricular filling pressures are elevated, thus decompressing the right ventricle while maintaining or improving left ventricular output. | {
"pile_set_name": "NIH ExPorter"
} |
Recently, there has been interest in the possibility that "differentiation-inducers" may have utility in the treatment of some malignancies. This concept is predicated on the belief that some malignancies are a result of a block in differentiation which if relieved would result in a more differentiated and therefore more benign condition. As a concept for therapy this approach holds the further promise that induction of differentiation could not only relieve the tumor burden but also increase the number of functional cells, that at least for some malignancies, an absence of is a major complication. HL60 has been a useful model system in the search for substances that are active as inducers of differentiation. HL60 is induced to differentiate to granulocyte-like cells by incubation with retinoic acid (RA), DMF, and DMSO or into monocyte/macrophage-like cells by incubation with 1,25-dihydroxyvitamin D3 and TPA. Of the many compounds that induce differentiation of HL60, RA has probably the most promise of being of use in the clinic. To the extent that it is possible, results in vitro should suggest treatments in vivo. To this end we have studied the differentiation effect of RA in nude mice carrying a transplantable HL60 tumor. There was no increase in life-span of treated animals and no evidence that tumor cells were induced to differentiate. These results were even more surprising because the tumor cells, established in culture and designated HL60/MRI, were found to be 100-fold more sensitive to RA in vitro than the parent HL60. In addition, RA induces HL60/MRI to differentiate to monocytoid cells. RA binds to serum albumin in a specific manner with an equilibrium constant of association of 10-6M-1. Thus, even at pharmacological concent concentrations of 1 MuM all of the circulating RA is bound to albumin. This binding may be a major factor for the absence of antitumor activity of RA in vivo as serum and/or serum albumin inhibit RA induced differentiation of HL60 in vitro. | {
"pile_set_name": "NIH ExPorter"
} |
Summary This application is in response to the Funding Opportunity Announcement PAR-13-253 ?Resource-Related Research Projects for Development of Animal Models and Related Materials (R24)?. We have recently developed a powerful new approach, ?CRISPR-activation (CRISPR-a)?, that allows the over-expression of endogenous genes in a stage- and tissue-specific manner in Drosophila. Here, we propose to build a resource of Drosophila transgenic lines that will allow the systematic analysis of the phenotypes associated with overexpression of fly genes orthologous to genes known or suspected to be associated with human diseases. Genetic model systems such as Drosophila have been used extensively to model human diseases associated with reduction or loss of gene function. By contrast, diseases associated with gene amplification, micro-duplication, or other changes resulting in over-expression have been more challenging to model. Over-expression models would be extremely valuable, as they would provide for example a system for relatively rapid identification of genetic or chemical modifiers of associated phenotypes. To fill this gap, we propose to: Aim 1. Develop CRISPR-a model flies for diseases associated with gene amplification or duplication. We have identified 279 Drosophila genes that are orthologous to genes amplified in various cancers or other diseases, or duplicated in syndromes associated with chromosomal microduplications. For each, we will generate a corresponding fly stock with two sgRNAs that can be combined with an activating form of Cas9. Additional candidates will be solicited from relevant labs, clinicians and coordinated efforts throughout the grant period; Aim 2. Develop CRISPR-a model flies for conserved rate- limiting enzymes. Disruption of one or more metabolic pathways is a direct cause of many diseases, and has important impact on many diseases and/or their treatments. The most impactful disruptions to a metabolic pathway occur when a genetic or other perturbation affects the levels of a rate-limiting enzyme. We have identified 300 fly genes orthologous to 390 human genes encoding rate-limiting components of metabolic pathways and will generate transgenic lines for overexpression. Aim 3. Perform an initial characterization of over-expression models. To increase the value of the resource to the community, we will systematically evaluate the gross phenotypes associated with overexpression of the lines created in Aims 1 and 2. Results will be collected on our existing RSVP phenotype database, which will facilitate search and view of the data by the community. | {
"pile_set_name": "NIH ExPorter"
} |
p66Shc has recently emerged as a master regulator of reactive oxygen species (ROS) production and cardiovascular oxidative stress responses. While a role for p66Shc as an 'aging' gene that amplifies ROS generation in mitochondria and leads to measurable changes in lifespan in animal models is convincing, there is almost no information on p66Shc functions in cardiomyocytes. Preliminary studies in this application implicate p66Shc as a target of an adrenergic receptor pathway involving ROS and PKC that is localized to caveolae and regulates AKT-FOXO3a phosphorylation and anti-oxidant gene expression. We identify an effect of the G1q-dependent hypertrophy pathway to increase p66Shc expression and target p66Shc to mitochondria where it is predicted to increase ROS generation. We identify a role for protein kinase C-4 (PKC4) and PKC5 to influence p66Shc phosphorylation and localization. p66Shc regulation by PKC4 is particularly noteworthy, as PKC4 also participates in mitochondrial events that control ROS accumulation and apoptosis. Studies in this application will consider the role of p66Shc as a G1q-induced gene product that cooperates with PKC to influence the evolution of cardiac failure phenotypes. Specific aim I will identify p66Shc activation mechanisms and p66Shc signaling functions in cardiomyocytes. The goal is to determine whether p66Shc participates in ROS-regulatory signaling mechanisms in caveolae and mitochondria that influence growth and/or apoptosis. Most studies do not resolve the signaling functions of p66Shc versus the smaller p46/p52Shc isoforms (that do not regulate Redox signaling). Studies in Specific Aim II will use p66Shc knockout models, siRNA gene silencing, and adenoviral mediated overexpression strategies to resolve the specific functions of p66Shc (and critical determinants within the p66Shc protein structure) in mechanisms that regulate ROS production and cardiomyocyte growth/apoptosis. Studies in Specific Aim III will focus on the role of PKC4 to cooperate with p66Shc to control cardiomyocyte remodeling. These studies are based upon preliminary data showing that PKC4 is a p66Shc binding partner that influences p66Shc-S36 phosphorylation and p66Shc subcellular targeting. Finally, studies in Specific Aim IV will examine whether p66Shc gene silencing prevents the structural remodeling and functional deterioration that develops in selected models of cardiac hypertrophy/failure. Evidence that p66Shc contributes to the etiology and/or pathogenesis of cardiac dysfunction would provide the basis for future research focusing on p66Shc as an 'aging gene' that influences the evolution of cardiovascular disorders associated with oxidative stress (ischemic, diabetic, atherosclerotic, hypertensive, and hypertrophic heart disease). The long-term goal of such studies would be to develop novel therapeutic strategies that prevent p66Shc expression or interdict p66Shc function that afford cardioprotection. PUBLIC HEALTH RELEVANCE: Studies in this application focus on p66Shc, an adapter protein that acts as a master regulator of oxidative stress responses and life span in animal models. Based upon the growing awareness that oxidative stress contributes to the evolution of ischemic, diabetic, hypertensive, and/or atherosclerotic heart diseases (i.e., oxidative stress is important regardless of etiology) and our recent studies showing that cardiac hypertrophy leads to increased p66Shc expression and enhanced p66Shc function in cardiomyocytes, studies in this application will examine the role of p66Shc-dependent Redox-regulatory mechanisms in cardiomyocyte growth and apoptosis responses. The long-term goal is to expose novel molecular strategies that can be used to reduce p66Shc expression and/or interdict p66Shc actions to prevent or slow the natural history of clinically important cardiovascular disorders. | {
"pile_set_name": "NIH ExPorter"
} |
Human plasmacytoid dendritic cells (pDC) constitute a rare subset of blood dendritic cells (DC), distinct from myeloid CD11c+, "conventional" DC (cDC). Through production of high levels of type I IFN in response to virus infection, pDC serve as a critical link between innate and adaptive antiviral immune responses. Recent observations from my laboratory have highlighted their particular role in the immune regulation of HIV-1 infection. pDCs, but not cDCs, undergo activation following CD4 mediated endocytosis of HIV-1 and subsequent activation of TLR7 with genomic RNA. Activated pDCs upregulate costimulatory molecules, produce pro-inflammatory cytokines and chemokines and activate immature cDCs in a bystander fashion. As a counterpoint to the induction of these anti-viral responses, HIV-activated pDCs simultaneously induce the differentiation of Tregulatory cells (Tregs) from naive resting CD4+ T cells, in a TLR7 dependent manner. Treg generation requires the expression of indolamine 2, 3-dioxygenase (IDO), an enzyme that catabolizes tryptophan to kynurenine, as it is reversed upon addition of the specific inhibitor 1 methyl-tryptophan. The T regs generated ("inducible T regs") inhibit the proliferation of activated T cells and maturation of cDC, thereby attenuating the induction of ongoing adaptive immune responses. Thus pDCs inhibit viral replication and promote anti-viral immunity, but at the same time limit the extent of immune activation. This newly ascribed property of pDCs is especially relevant in HIV infection where control of excessive immune activation could be essential to prevent virus dissemination and progression of disease. In this application we propose to: (1) Determine the role of TLR7 and TLR9 in HIV-induced activation of IDO;(2) Elucidate the players in the non- canonical NF: B pathway for IDO expression;(3) Examine the pDC-derived products that cause T reg generation. These studies will greatly improve our understanding of the events that follow pDC activation by HIV and potentially result in clinically applicable approaches to enhance anti-HIV immune responses in vivo. PUBLIC HEALTH RELEVANCE: These studies will investigate the mechanisms used by the HIV virus to evade the immune system. Specifically, we will explore how HIV induces the generation of T cells which have suppressive qualities and block the development of several beneficial anti-viral responses. By understanding how these suppressive T cells are generated we can devise strategies to modulate their function and promote anti-viral immunity. | {
"pile_set_name": "NIH ExPorter"
} |
The Section on Organelle Biology investigates the global principles underlying secretory membrane trafficking, sorting and compartmentalization within eukaryotic cells. Live cell imaging of green fluorescent protein (GFP) fusion proteins in combination with photobleaching and photoactivation techniques are being used to investigate the subcellular localization, mobility, transport routes and binding interactions of a variety of proteins with important roles in the organization and regulation of membrane traffic and compartmentalization. Quantitative measurements of these protein characteristics are used in kinetic modeling and simulation experiments in order to test mechanistic hypotheses related to protein and organelle dynamics. Among the topics currently under study include: growth and maintenance of endoplasmic reticulum (ER) and Golgi morphology in mammalian cells and in developing Drosophila embryos; the mechanism(s) of secretory protein transport into and out of the Golgi apparatus; membrane binding/dissociation kinetics of trafficking machinery and its regulation; the generation and maintenance of cell polarity; and, organelle breakdown and reassembly during mitosis. We have also recently developed a photoactivatable GFP, whose mechanism of photoactivation is currently being investigated. Development of a Photoactivatable GFP Photoactivation is the rapid conversion of photoactivatable molecules to a fluorescent state by intense irradiation and is a useful method for marking and monitoring selected molecules within cells. Previous efforts to develop a photoactivatable protein capable of high optical contrast when photoactivated under physiological conditions have had limited success. By mutagenizing wild type GFP (wtGFP), we have developed a variant of GFP that allows selective marking of proteins through photoactivation. The wtGFP normally exists as a mixed population of neutral phenols and anionic phenolates producing its major 397 nm and minor 475 nm absorbance peaks, respectively. Upon intense illumination of the protein with ultraviolet or ~400 nm light, the chromophore population undergoes photoconversion and shifts predominantly to the anionic form, giving rise to an increase in minor peak absorbance. This produces a ~3-fold increase in fluorescence upon excitation at 488 nm. The photoactivatable variant of wild type GFP that we developed (called PA-GFP for ?photo? and ?activatable?) contains a histidine substitution at the T203 position of GFP that results in its having a negligible minor absorbance peak. Photoconversion with ~400 nm irradiation produces a large increase in absorbance at the minor peak and thus a more noticeable optical contrast under 488 nm excitation. Upon photoactivation of living cells, PA-GFP exhibited an optical enhancement of nearly two orders of magnitude, making it suitable to mark specific protein or cell populations. The speed with which an optical signal was obtained and the absence of signal from newly synthesized proteins, furthermore, indicated PA-GFP photoactivation was a preferable labeling method to photobleaching for studying the temporal and spatial dynamics of proteins. We used the photoactivatable GFP both as a free protein to measure protein diffusion across the nuclear envelope and as a chimera with a lysosomal membrane protein to demonstrate rapid interlysosomal membrane exchange. Our results suggest that the photoactivatable variant of GFP, PA-GFP, has the potential for addressing many fundamental questions in cell and developmental biology. Dissection of COPI dynamics in vivo Cytosolic coat proteins that bind reversibly to membranes have a central role in membrane transport within the secretory pathway. One well studied example is COPI or coatomer, a heptameric protein complex that is recruited to membranes by the GTP-binding protein Arf1. Assembly into an electron-dense coat then helps in budding off membrane to be transport between the ER and Golgi. To study COPI dynamics in vivo, variants of GFP were fused to the carboxy terminus of the eCOPI subunit of coatomer and expressed in ldlF cells, which contain a mutated eCOP that at 40oC is degraded, causing coatomer inactivity and growth inhibition. The cells grew indefinitely at 40oC, indicating that eCOP-GFP could substitute functionally for endogenous eCOPI in coatomer complexes in these cells. The identity and behavior of coatomer-containing membranes, assessed by eCOP-YFP labeling, were studied in dual-color time-lapse experiments in ldlF cells co-expressing cyan fluorescent protein (CFP)-tagged secretory cargo markers. eCOP-YFP was present on juxtanuclear Golgi membranes and on pre-Golgi transport intermediates containing secretory cargo, and it was depleted from retrograde (Golgi-to-ER) transport intermediates. The pre-Golgi intermediates remained brightly labeled with eCOP-YFP as they tracked into the Golgi. To gain insight into the role of COPI on anterograde (ER-to-Golgi) transport intermediates we used photobleaching techniques to investigate the characteristics of COPI binding and release from membranes. Upon photobleaching either Golgi or pre-Golgi structures expressing eCOP-GFP in ldlF cells, fluorescence recovered onto these structures exponentially with a half-time of 35 s. This indicated that coatomer binding and dissociation from membranes occurs continuously. To test whether this activity is coupled to vesicle budding, we measured the kinetics of coat exchange on and off membranes at low temperatures, in which vesicle transport is nonexistent. We found that at all temperatures down to 4oC, eCOP-GFP underwent binding and release from Golgi membranes, with no abrupt change in the kinetics on reaching temperatures at which vesicle budding is slowed or inhibited. The data thus indicated that the cycling of coatomer on and off membranes can be uncoupled from vesicle formation, and that feedback from productive vesicle budding is not necessary for COPI dissociation. Based on these findings, we proposed that membrane binding and release of COPI serves to initiate and stabilize lateral sorting and segregation of cargo into membrane domains that progressively differentiate into pleiomorphic membrane transport intermediates and/or vesicles for membrane transport in the ER/Golgi system. Arf1 regulation of COPI dynamics COPI is known to be recruited to membranes by the GTP binding protein Arf1 and GTP hydrolysis of GTP-bound Arf1 is believed to be necessary for COPI release from membranes. To gain further insight into Arf1 regulation of COPI dynamics, we asked if Arf1 GTP hydrolysis is sufficient for COPI to dissociate from membranes; that is, do Arf1 and COPI dissociate together from membranes. To address this question, we utilized a functional Arf1-CFP chimera that allowed us to visualize the behavior of Arf1 and coatomer in the same cell. FRAP experiments revealed the half-time for Golgi membrane dissociation of Arf1-CFP (13s) was significantly faster than for eCOP-YFP (30 s) suggesting Arf1 and coatomer dissociate independently from Golgi membranes. This was supported by results from experiments using BFA, which inhibits Arf1 and COPI recruitment to membranes, allowing the dissociation of these proteins to be observed in the absence of rebinding. On treatment with BFA, Arf1-CFP dissociated significantly faster (T1/2 13 s) than eCOP-YFP (t1/2 30s). These findings indicated, therefore, that Arf1 and coatomer dissociation from Golgi membranes are regulated differently. A mathematical formulation of COPI and Arf1 membrane binding and dissociation kinetics was able to fit simultaneously both Arf1 and COPI photobleaching data and the Arf1 and COPI release kinetics following BFA treatment, providing new quantitative estimates of the lifetime of these molecules on membranes and their bi | {
"pile_set_name": "NIH ExPorter"
} |
This 2-stage, Phase I/II trial evaluates the safety and efficacy of HP228 administered by subcutaneous injection to advanced stage cancer patients receiving cisplatin, doxorubicin or paclitaxol as palliative chemotherapy. HP228 is a cytokine-restraining agent with a broad spectrum of anti-inflammatory, analgesic and antipyretic properties, which may attenuate levels of TNF-alpha, IL1b, IL-6, and may decrease certain treatment-related toxicities. The trial is completed. | {
"pile_set_name": "NIH ExPorter"
} |
This project is designed to further purify and characterize various forms of glutamate decarboxylase (GAD), cysteic/cysteinesulfinic acids decarboxylase (CAD/CSAD) and serine-transhydroxylmethylase (serine-THM), the biosynthetic enzymes for GABA, taurine and glycine respectively, and to delineate the role of these enzymes in the regulation of the effective levels of GABA, taurine and glycine in connection with their important roles as neurotransmitters or modulators in mammalian central nervous system. The purified enzyme preparations will also serve as antigens for the production of specific antibodies, so that the GABAergic neurons and those neurons containing CAD/CSAD and serine-THM can be identified at cellular and subcellular levels by immunocytochemical methods. In addition, I also plan to perform the following studies (1) To purify the high molecular weight (MW) GAD from the cell body-enriched fraction and to determine whether the high MW GAD represents as a precursor or as a higher polymeric form of low MW GAD, (2) To elucidate the mechanism of conversion of high MW GAD to low MW GAD with special emphasis on the nature of the converting factor(s), (3) To make monoclonal antibodies against various forms of GAD (high and low MW GAD and glial GAD), CAD/CSAD and serine-THM so that sufficient quantities of antibodies will be available for immunochemical and immunocytochemical characterization of various neuronal pathways involving GABA, glycine and taurine as transmitters or modulators, (4) To elucidate the function of glial GAD and GAD in non-neuronal tissues, e.g., kidney and heart. | {
"pile_set_name": "NIH ExPorter"
} |
The HIV-1 envelope (env) glycoprotein (gp120-gp41) plays critical roles during virus entry into the target cell mediating binding to cellular receptors and the fusion of viral and cellular membranes. The biologically relevant form of env is a heavily glycosylated oligomeric complex. The location of th eenv on the viral membrane renders it an important target for vaccine development. Indeed, it is the only viral component accessible to antibodies and is the viral antigen to which neutralizing antibodies are directed. Unfortunately, most neutralizing antibodies to env exhibit limited cross-reactivity with other HIV-1 isolates, notaly, primary isolates. However, that much more broadly neutralizing antibodies to conformation- dependent epitopes canbe developed has been indicated, and an effective vaccine should elicit potent, broadly neutralizing antibodies to a number of epitopes. Thus, it is important to understand the determinants in env that stimulate the production of antibodies that are broadly reactive. We have found that immunizationwithsoluble, oligomeric env (BH8) effectively generates monoclonal antibodies (Mabs) that are broadly cross- reactive. The majority of these Mabs reacted with conformational epitopes, many reacting preferentially or exclusively with env oligomer. Several novel neutralizing Mabs to oligomer dependent, and specific,epitopes in the fp41 ectodomain were also identified. These studies have indicated the importance of investigating the oligomeric antigenic structure of env from primary isolates. In addition, evaluation of glycosylation contribution in epitope masking on oligomeric env will be important in determining if simple changes can be made to enhance cross-reactive neutralizing antibody production. One way neutralizing antibodies could work is to perturb some betastable property of env required in fusion. This kind of activity could inactivate virus andplay a role in preventing cell-cell transmission. It is like that these antibodies willreact with epitopes that are dependent on oligomeric structure andreact with highly conserved determinants. The overall goal of this project is to fully evaluate the consequences of env oligomerization and glycosylationon antigenic structure in primary isolates, and to understand how oligomeric env modulates during interactions with cellular receptors. Specifically, we will: 1) Develop and characterize soluble oligomeric and glycosylation-altered envs from primary HIV-1 isolates; 2) Assess the effects of oligomerization and glycosylationon antigenic structure in small animals, developing Mabs, and identify highly conserved and neutralizing epitopes; and 3) Characterize the env-mediated fusion process using a novel video fluorescence microscopy technique - monitoring receptor-induced conformation changes in situ in env, analyzing these changes with a variety of exisitng and newly developed Mabs as probes, and identify important functional domains as env interacts with cellular receptors. | {
"pile_set_name": "NIH ExPorter"
} |
The nickel enzyme CODH (carbon monoxide dehydrogenase) plays a critical role in carbon cycling via microbial metabolism that converts CO2 to cellular carbon and, as such, is essential to life on this planet. Functional model complexes are lacking and thus this proposal aims to prepare heterobimetallic complexes with Ni/Fe centers resembling CODH with the goal to answer questions regarding the enzyme function and ultimately design systems for selective and efficient CO2 reduction. Purely functional models, those that are electrocatalysts for CO2 reduction, contain only one type of metal ion and operate at large overpotentials. New synthetic CODH model complexes are needed to illuminate the details necessary for producing a structurally faithful functional model. Our strategy involves the synthesis of heterobimetallic Ni/Fe complexes for CO2 reduction, a first in synthetic chemistry. We will utilize functionalized diimineo-dioxime ligands that allow sie-selective incorporation of two distinct metal ions. Questions to be addressed with these model complexes include: how does CO2 bind to heterobimetallic Ni/Fe centers? How does the Lewis acidic metal ion affect CO2 binding? What is the binding equilibria and extent of activation at reduced Ni/Fe centers? What conditions favor electrocatalytic reduction of CO2? Can the presence of a Lewis acidic metal improve Faradaic efficiencies and/or lower the overpotential necessary for catalysis? The Peters lab is suited to conduct these studies. Prof. Jonas Peters is an expert in synthetic organometallic and inorganic chemistry. Additionally, he is knowledgeable in the field of electrochemistry and electrocatalytic methods. Finally, the academic environment and facilities available at Caltech encourages rich collaborative discussions that will ensure the success of the proposed studies. | {
"pile_set_name": "NIH ExPorter"
} |
Primary Hyperparathyroidism is described best today as a relatively asymptomatic disease characterized by mild elevations in the serum calcium and parathyroid hormone concentrations. Over the past 18 years, this research project has helped to define the clinical, biochemical, densitometric and histomorphometric features of primary hyperparathyroidism in the modern era. As a major source of new insights, the investigation has helped to elucidate aspects of this disease that were not previously appreciated. The overall objective of this research project remains the definitive description of extent, course, and reversibility of the manifestations of primary hyperparathyroidism. Other important challenges will be pursued in order to achieve a more complete understanding of primary hyperparathyroidism. The following Specific Aims will be pursued: (1) to define the natural history of primary hyperparathyroidism, and the reversibility of its manifestations, over a uniquely long, systematic 23-year period of investigation. This effort will include characterization of new areas of inquiry in neuropsychological function, histomorphometric, geometrical, and cellular aspects of bone in primary hyperparathyroidism; (2) to characterize a newly recognized presentation of asymptomatic primary hyperparathyroidism without hypercalcemia; (3) to complete the characterization of the bone remodeling unit in primary hyperparathyroidism; 4) to determine fracture risk in primary hyperparathyroidium. This project will utilize state-of-the-art techniques, including bone densitometry, peripheral quantitative tomography, bone histomorphometry, quantitative backscattered electron imaging, quantitative assessment of vertebral fractures, and cognitive assessment. By the end of the renewal period, we expect to have fully characterized the long-term natural history of hyperparathyroidism, with or without surgery, as well has to have defined features of this disease that have, up to now, eluded successful investigation. The results should lead to the establishment of rational recommendations for the management of primary hyperparathyroidism. | {
"pile_set_name": "NIH ExPorter"
} |
Project Summary The human immunodeficiency virus type 1 (HIV-1) is a retrovirus that completes its viral life cycle when the newly assembled virion is released from the infected cell. The HIV-1 structural protein, Gag, recruits the host cell?s endosomal sorting complex required for transport (ESCRT) proteins towards the neck of budding virions to sever the nascent viral particle from the plasma membrane. In addition to viral-like particle (VLP) release, ESCRTs are utilized in membrane scission of other topologically equivalent cellular processes such as vesicular trafficking, cell division, exosome biogenesis, and plasma membrane repair. Recent advances in molecular biology and virology highlight the importance and timing of recruitment of ESCRTs in achieving proper viral particle release. However, a description of the biophysical mechanism of ESCRT-mediated scission during HIV release remains a major goal in the field. Newly developed methods in our laboratory have now made possible the encapsulation of functional human ESCRT and Gag proteins inside giant unilamellar vesicles (GUVs), thereby recapitulating the correct topology for ESCRT function in vitro. We have integrated a high-speed confocal microscope with optical trapping capabilities which allows the visualization and investigation, with piconewton resolution, of the force generated during scission of a single membrane neck. Control of the reaction is modulated by UV photolysis of a caged-nucleotide for this ATP-dependent process. Together, these innovations have given us a unique ability to interrogate HIV-1 release by ESCRTs under a biophysical lens. This proposed research represents a focused and innovative approach to investigate the ATP-dependent membrane scission mechanism of human ESCRT proteins in the setting of HIV-1 release; directly building and expanding on our previous success with the yeast ESCRT system. Specifically, in aim 1, I will identify the scission mechanism of human ESCRTs. Subsequently, aim 2 will provide a biophysical explanation of the interplay between Gag binding and membrane scission by the ESCRT machinery. The overarching hypothesis for this proposal is that ESCRTs apply mechanical force to membrane necks which destabilizes them and leads to their scission. Ultimately, successful completion of this work allows for a detailed biophysical understanding of HIV-1 release by ESCRTs that may lead to the design of novel antivirals. | {
"pile_set_name": "NIH ExPorter"
} |
In many genetically based diseases specific mutations or deletions can result in the loss of biological activity of a protein whose function is essential for the normal lifestyle of the cell. Often times, the particular mutation results in a failure of the protein to properly fold and therefore not be delivered to its site of action inside the cell, or secreted out of the cell. It has now been established that the pathway of protein folding is dependent upon the participation of a class of proteins now referred to as molecular chaperones. While not conveying any direct information for the folding process, molecular chaperones help reduce the possibility of a protein undergoing a nonproductive folding pathway which could lead to its misfolding and/or aggregation. Proteins unable to achieve a stable folded conformation, due for example to a specific mutation, are recognized by and retained by the actions of one or members of the molecular chaperones. Thus, molecular chaperones have been suggested to act as a type of "quality control" system inside the cell, facilitating the maturation of most polypeptides but retaining those unable to adopt their biologically active conformation, likely targeting the latter for eventual degradation. Here, we will continue with our studies examining the early events of protein synthesis, and the role of different molecular chaperones in facilitating protein maturation. Particular attention will be devoted to identifying cellular components, including the hsp70 chaperone, which interact with nascent polypeptides undergoing synthesis on the ribosome. Two potential co-factors we suspect are involved in the ATP dependent reaction cycle of hsp70 with nascent and newly synthesized proteins will be identified and characterized. In a series of related studies, we will examine the role of different molecular chaperones in the synthesis of alpha and beta tubulin and their assembly into the alpha/beta heterodimer. We suspect that newly synthesized alpha and beta tubulin initially interact with the hsp70 chaperone, and then are transferred over to another chaperone, the so- called TCP-1 chaperonin, where folding and assembly of the 65 tubulin heterodimer commences. In addition, we will follow up on our studies indicating that the assembly of 6S tubulin into microtubules, either in vitro or in vivo, again involves the participation of one or members of the chaperone family. We anticipate that these studies will further define the critical roles played by molecular chaperones in the complex pathways by which cellular proteins achieve their biologically active conformation inside the cell. | {
"pile_set_name": "NIH ExPorter"
} |
This is a continuing investigation of the factors controlling survival of pathogenic bacteria and certain fungi on human skin. We demonstrated that certain pathogenic bacteria and Candida albicans, when deposited on human skin and partially occluded, are significantly reduced in number or even eliminated. We further observed that the antimicrobial activity of skin varies from individual to individual; some subjects tend to destroy these applied bacteria while others do not. This antimicrobial property of skin is effective against Staphylococcus aureus and Candida albicans. Skin lipids have been considered antimicrobial by some authors. We have delineated populations of men in terms of persistence or inhibition of S. aureus applied to their skin. The normal flora of these polar-type populations will be quantified. It will be determined whether these individuals tend to be at the high or low ends of a distribution curve in terms of the number and types of their normal skin flora. We will correlate the presence of antimicrobial skin lipids with the absence of pathogenic bacteria. Skin lipids will be extracted with acetone and fractionated. Both quantitative and qualitative analyses of skin lipids in terms of antimicrobial activity will be performed. Different fractions of lipid will be tested for their ability to prevent multiplication of microorganisms. These fractions will be characterized chemically. The isolation and successful identification of these fractions will be useful in investigating the effectiveness of these antimicrobial agents in the prevention of skin infection. | {
"pile_set_name": "NIH ExPorter"
} |
Studies of Photosensitized Oxidation of model biological systems are proposed. The role of singlet oxygen, superoxide ion, and other reactive intermediates in the oxidation of amino acids, enzymes and nuclear bases will be determined. The mechanism of protective action of carotenes, amines, and phenols will be studied using kinetic techniques, flash photolysis, and competitive inhibition with specific singlet oxygen and superoxide traps. Products will be determined. The chemistry of superoxide ion will be further elucidated, as will the chemistry of metal-oxygen complexes. Models for enzymic oxidations should be clarified by these studies, and a possible carcinogenesis mechanism can be tested. BIBLIOGRAPHIC REFERENCES: Ta-Yen Ching and Christopher S. Foote, "Chemistry of Singlet Oxygen. XXII. Photooxidation of Nitrones," Tetrahedron Lett., 3771 (1975). Christopher S. Foote, "Photosensitized Oxidation and Singlet Oxygen: Consequences in Biological Systems," in Free Radicals in Biology, II, 85 (1976), Edited by W. A. Pryor, Academic Press, New York. | {
"pile_set_name": "NIH ExPorter"
} |
We propose to develop new therapeutic modalities for human tumors using as a model human malignancies heterotransplanted in nude mice. In particular, we propose: 1) to develop fast and reliable assays for testing the effectiveness of drugs and other therapeutic modalities (hormones, ionizing radiations and hyperthermia) against human tumors. In addition to improving the subcutaneous and intrarenal systems, we are investigating the intracerebral system which presents unique possibilities for studies of the blood-brain barrier permeability to certain drugs and the anterior chamber of the eye which offers the possibility to perform rapidly microtests of drug sensitivity on minimal amounts of tumors; 2) to study the predictivity of human tumors heterotransplants for clinical therapy by a) testing known antitumor agents or combinations of them against individual human tumors and comparing the effect of the same agent(s) against the same tumor in the nude and in the patient of origin; b) testing the statistical predictivity by means of tumor panels. We will develop panels of 14 tumors each for lung carcinoma, carcinoma of the uterus and carcinoma of the ovary (as we already did for colon and breast carcinomas and melanomas) and test nine known antitumor drugs against all 42 tumors; 3) to investigate the effect on human tumors of drugs which have a short biological half life and have to be continuously administered. We have developed a highly efficient system for the continuous infusion of drugs subcutaneously and intraperitoneally in nude mice. Using such a system, we are in the process of studying the effect of Thymidine and have started the study of hydroxyurea and of Ara C. | {
"pile_set_name": "NIH ExPorter"
} |
Diabetes is a serious disease affecting over 1 million people in the United States. The standard treatment, injection of insulin, is not sufficient to control blood glucose in many patients and leads to complications such as kidney failure, limb amputations and blindness. Recently a new experimental therapy has shown promise of cure for this disease. The therapy consists of isolating clusters of the insulin secreting cells, called islets from cadaver pancreases and transplanting them into diabetic patients. While the results of these experimental transplants have been promising, it has been difficult to keep the islets alive once they have been isolated from the pancreas. Recently it has been shown in an animal model of diabetes, that culturing islets in a new type of bioreactor system invented at NASA, can significantly improve the survival of islets outside the body and improve the success of islet transplantation. The goal of this project is to adapt this culture system to culture human islets so that they will improve the outcome of islet transplantation to treat diabetes. [unreadable] [unreadable] Project Narrative: Pancreatic islet transplantation holds great promise for a cure for type 1 diabetes. We have experimental evidence that culturing islets in the Rotary Cell Culture System can improve the survival and function of islets after transplantation. The goal of this proposal is to verify the previous results in human patients so that this technology can be used to improve this promising therapy for diabetes. [unreadable] [unreadable] [unreadable] | {
"pile_set_name": "NIH ExPorter"
} |
The mission of this Program is to prepare a cadre of outstanding cancer epidemiologists through rigorous academic training in research methodology and the epidemiology and biology of cancer, and/or mentored research. Those in the Molecular & Genetic Epidemiology Sub-program receive additional training in the application of molecular genetic techniques to epidemiologic research. A distinguished faculty provides substantial opportunity for research experience. There are three types of trainees: Doctoral Candidates in Epidemiology include both pre-doctoral candidates and those holding a prior doctoral degree (generally clinicians) who undertake 2-3 years of coursework in addition to research; Clinical Researchers include physicians with specialty training in cancer preparing for careers in clinical epidemiology who undertake a one year Master's degree program in methodology followed by a year of research; and Post-doctoral Fellows who hold a prior doctora! degree in Epidemiology or in Biology and undertake mentored research (and coursework if needed) in preparation for an academic career. Trainee positions are generally awarded for 2 or 3 years. The number of trainees requested include 3-4 pre-doctoral and 1-2 post-doctoral Doctoral Candidates; 2 Clinical Researchers; 2 Post-doctoral Fellows with an additional 4 pre-doctoral Doctoral Candidates and 3 Post-doctoral Fellows in the Sub-program for a total of 16 trainees. The Program is based in the Department of Epidemiology of the Harvard School of Public Health. A dedicated molecular epidemiology laboratory is available. This proposal continues a long history of excellence in training in cancer epidemiology at Harvard University. | {
"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"
} |
ProjectSummary Thegoalofthiscompetitiverenewalapplicationistoprovideavarietyofcomplementarytrainingopportunities indentalandcraniofacialresearchattheUniversityofIowa(UI)andtodevelopacohortofbasicresearchers anddentalscholarswhoseinvestigativeskillswillmeetthechallengesoforalhealthresearchforthe21st century.Theaimsaretopreparetraineesformeaningfulcareersinoralhealthresearchandacademicsby providingoutstandinginterdisciplinarydidactictrainingandrigorousresearchmentoringexperiences emphasizingongoingreviewandcritique.Theproposedprogramcontinuation,buildingon30+yearsof researchtrainingsuccess,hasafocusondentalgraduates,butrecognizesthatbasicscientistsalsoplay importantrolesinoralhealthresearch;?thus,itwillalsooffertrainingopportunitiesfornon-dentists.TheR90 componentreflectstheincreasinglyimportantrolethatinternationaldentistshave,andwillcontinuetohave,in ouroralhealthresearchprograms.Thefivemajorresearchareasinwhichtrainingisoffered,1) Bioengineering,TissueEngineering,StemCells,Biomaterials,andMaterialsResearch2)Immunology, Inflammation,Microbiology,Caries,andMicrobiomeResearch3)Craniofacial,OralBiology,Genetics,and DentalDevelopmentResearch4)PublicHealth,EpidemiologyandBehavioralSciences,and5)OralHealth PolicyResearch,reflectthestrengthsoftheCollegeofDentistry,thehealthsciencecolleges,UIinstitutesand centersandrangeacrossthefulltranslationalspectrum.Specifically,weoffertrainingfordentistsandnon- dentiststopursueaPhDinOralSciences(orinoneofthesixotherPhDprogramsfromwhichwewillrecruit, asappropriate),ortopursuepost-doctoraltraining.Inaddition,therewillbeanopportunitytoobtaina CertificateinTranslationalandClinicalInvestigationundertheauspicesoftheUIInstituteforClinicaland TranslationalSciences(ourCTSA-fundedinstitute).Agroupof28experiencedandtalentedprogramfaculty fromavarietyofdisciplinesisavailabletomentortrainees.Trainingwillbesupervisedbyanexperienced DirectorandAssociateDirector,assistedbyaFacultyLeadershipteam.Theprogramwillhaveinternaland externaladvisorycommittees,anewly-establishedGrant-writingCommittee,anewly-establishedFaculty MentoringProgram,andaCommitteeonRecruitmentandDiversity.Insummary,theprogrambringstogether productive,well-fundedmentorswithpre-andpost-doctoraltraineesinanenvironmentwithastrong institutionalcommitmenttodevelopoutstandinginterdisciplinaryresearchtraining,astronginstitutionalrecord oftraininginrelevantdisciplines,andexcellenteducationalandresearchfacilities.Theoutcomewillcontinue tobethedevelopmentofacadreoforalhealthresearchersequippedtofunctionatthecuttingedgeoftheir disciplines. | {
"pile_set_name": "NIH ExPorter"
} |
The relationship between a primary sequence and the three-dimensional structure is a fundamental issue in macromolecular folding. RNA folding is governed by rules that represent an illuminating contrast to the more popular "protein folding problem." RNA folding involves many metastable intermediates that are prerequisite for folding to the native structure. Previous studies, primarily ensemble measurements, demonstrated that elucidation of the structural and dynamic characteristics of these intermediates is crucial for understanding RNA folding. Ensemble measurements, however, measure average properties and often lack sufficient resolution to obtain detailed mechanistic insights involving folding intermediates. Because the ensemble generally populates several intermediate states under a variety of useful folding conditions, ensemble measurements only report an average behavior of these overlapping populations. This is clearly deleterious to; for example, establishing microscopic rates of reaction between the various states in the folding pathway. We will conduct single-molecule folding studies of two homologous ribozymes of abut 260 residues whose ensemble-determined folding pathways consist of defined, but overlapping intermediates and are free of kinetic traps. Folding will be monitored by fluorescence resonance energy transfer between two fluorophores attached at specific locations in the RNA. Aim 1 is to characterize the distribution and structural fluctuations of folding intermediates and to compare them with structural fluctuations during folding. Aim 2 is to identify fluctuations and dynamics among subdomains of the large ribozyme from Bacillus subtilis RNase P with strategic FRET labeling. Aim 3 is to compare folding of mesophilic and thermophilic homologues. These results will elucidate folding pathways and their associate intermediates and address how sequence affects the energetics and structures of folding intermediates. | {
"pile_set_name": "NIH ExPorter"
} |
Fanconi anemia (FA) is a recessively inherited disease characterized by congenital defects, bone marrow failure, and cancer susceptibility. The disease has also been considered by some as a segmental progeriod entity, as FA patients develop a range of tissue-specific premature onset and accelerated aging phenotypes, including bilateral cataracts, skin atrophy, reduced muscle mass, premature ovarian failure, higher frequency of osteoporosis and osteopenia, and diabetes. Fifteen genes have now been described that are mutated to cause FA, three discovered by our group. Recent evidence suggests that FA proteins function in a DNA damage response pathway involving the proteins produced by the breast cancer susceptibility genes BRCA1 and BRCA2. A key step in that pathway is modification of two FA proteins, FANCD2 and FANCI. The modification, monoubiquitylation, results in redistribution of FANCD2-FANCI to specific spots in the nucleus where BRCA proteins also localize. When we initiated this project in 2001, five FA proteins (FANCA, -C, -E, -F, and -G) were found to interact with each other to form a multiprotein nuclear complex, the FA core complex. This complex functions upstream in the pathway and is required for FANCD2-FANCI monoubiquitylation. We purified the FA core complex and found that it contains at least seven new components in addition to the five known FA proteins. We have characterized these new components and shown that they are important for the FA-associated DNA damage response pathway, as summarized below. One new component of the FA core complex, termed PHF9, possesses ubiquitin ligase activity in vitro and is essential for FANCD2-FANCI monoubiquitylation in vivo. PHF9 is defective in a group of Fanconi anemia patients, and therefore represents a novel Fanconi anemia gene (FANCL). Our data suggest that PHF9 plays a crucial role in the Fanconi anemia pathway as the catalytic subunit for monoubiquitylation and FANCD2-FANCI. The discovery of PHF9/FANCL might provide a potential target for new therapeutic modalities. We then showed that the 95 kd subunit of the Fanconi anemia core complex is defective in FA complementation group B patients (the gene is named FANCB). FANCB is X-linked and present in only one active copy. Thus, FANCB could represent a vulnerable target in the machinery that maintains genome stability, because it will take only one mutation to inactivate FANCB in males, compared to two mutations required to inactivate other autosomal FA genes. We demonstrated that the 250 Kda subunit of the FA core complex is mutated in FA patients of a new complementation group, FA-M. The gene was named FANCM. FANCM has a conserved helicase domain and a DNA remodeling activity. FANCM has at least three important roles in the FA DNA damage response pathway. First, it plays a structural role, allowing assembly of the FA core complex, because in its absence, the nuclear localization and stability of several FA proteins are defective. Second, FANCM translocates and remodels various DNA structures, which are important for subsequent DNA repair. Third, FANCM is hyperphosphorylated in response to DNA damage, suggesting that it may serve as a signal transducer through which the activity of the core complex is regulated. We have identified the 100 Kda subunit of the FA core complex, FAAP100, and shown that this protein is required for stability and a key function of the complexmonoubiquitination of FANCD2-FANCI. We have identified the 24 Kda subunit of the FA core complex, termed FAAP24, which forms a heterodimer with FANCM. FAAP24 can recognize structured DNA that mimics intermediates generated during DNA replication. Moreover, it can target FANCM to such structures. Cells depleted of FAAP24 show phenotypes that are characteristics of FA cells. Our results demonstrate that FAAP24 is an integral component of the FA core complex, We also collaborated with other labs to demonstrate that PALB2, a partner of BRCA2, is the gene defective in Fanconi anemia complementation group N patients. In further mechanistic studies, we demonstrated that FANCM possesses an ATP-independent binding activity and an ATP-dependent bi-directional branch-point translocation activity on a synthetic four-way junction DNA, which mimics intermediates generated during homologous recombination or at stalled replication forks. We found that the ATP-dependent activities of FANCM are required for cellular resistance to a DNA crosslinking drug, mitomycin C (MMC), but not for the monoubiquitination of FANCD2-FANCI. In contrast, monoubiquitination requires the entire helicase domain of FANCM, which has both ATP- dependent and independent activities. These data are consistent with participation of FANCM and its associated FA core complex in the FA pathway at both signaling through monoubiquitination and the ensuing DNA repair. We identified two new components in the FA core complex, MHF1 and MHF2. These two proteins form a histone-fold heterodimer that associates with FANCM to form a DNA-remodeling complex conserved from yeast to human. MHF stimulates DNA binding and replication fork remodeling by FANCM. In the cell, FANCM and MHF are rapidly recruited to forks stalled by DNA interstrand crosslinks, and both are required for cellular resistance to such lesions. In vertebrates, FANCM-MHF associates with the Fanconi anemia (FA) core complex, promotes FANCD2 monoubiquitination in response to DNA damage, and suppresses sister-chromatid exchanges. Yeast orthologs of these proteins function together to resist MMS-induced DNA damage and promote gene conversion at blocked replication forks. Thus, FANCM-MHF is an essential DNA-remodeling complex that protects replication forks from yeast to human. We showed that another FA protein, FANCJ, becomes hyperphosphorylated in response to DNA damage. We are investigating if this phosphorylation regulates activity of FANCJ in DNA repair. We collaborated with Dr. Lei Li's lab to develop a chromatin-IP-based strategy termed eChIP and elucidated how various FA proteins are recruited to the interstrand DNA crosslinks that block replication. We found that BRCA-related FA proteins (BRCA2, FANCJ/BACH1, and FANCN/PALB2), but not FA core and I/D2 complexes, require replication for their crosslink association. FANCD2, but not FANCJ and FANCN, requires the FA core complex for its recruitment. FA core complex requires nucleotide excision repair proteins XPA and XPC for its association. Thus, FA proteins participate in distinct DNA damage response mechanisms governed by DNA replication status. We have recently identified FAAP20 as a new component of the FA core complex. FAAP20 contains an ubiquitin binding motif and is required for FA core complex to localize to DNA damage sites. Depletion or inactivation of FAAP20 reduces monoubiquitylation of FANCD2-FANCI, indicating that FAAP20 is an important player of the FA pathway. The fact that FANCM and MHF1-MHF2 constitute a conserved DNA remodeling machine prompted us to perform crystal structure studies of this complex. So far, we have also solved the crystal structure of MHF1-MHF2 complex in conjunction with the region of FANCM that binds MHF. The structure reveals how these three proteins interact with each other to form a complex and recognizes DNA. We are continuing to identify and characterize additional components of the FA complex. The eventual goal is to analyze the full pathway of DNA crosslink repair, assess its role during normal aging, and in DNA repair-deficient diseases. | {
"pile_set_name": "NIH ExPorter"
} |
Oral administration of soluble protein antigens typically induces antigen specific systemic nonresponsiveness. However, the PI has found that when a model food protein, ovalbumin (OVA), is orally administered to mice infected with a nematode parasite (Heligmosomoides polygyrus), this normally tolerogenic stimulus primes for a systemic OVA specific Th2 response. We have shown that the polarized Th2 cytokine response induced by this enteric infection plays a central role in determining whether or not tolerance to OVA is induced. We have also found that H. polygyrus infection potentates the response to oral antigen by enhancing T cell clonal expansion and upregulating costimulatory molecule expression on mucosal antigen presenting cells (APC). In Specific Aim 1,we will further explore the mechanistic basis for this effect by examining the influence of infection on various APC populations which can present orally administered antigens in mucosal and peripheral lymphoid tissue. To examine the ways in which helminth infection alters the OVA specific T cell response, T cells from OVA specific TCR transgenic mice will be adaptively transferred into normal BALB/c mice and their response will be tracked in vivo. In addition to providing insight into the ability of an infection to bias the response to other antigens, these experiments will provide basic information on antigen presentation in the mucosal immune system and the means by which it orchestrates decisions concerning tolerance and immunity. Since parasitic infection is endemic in many parts of the Third World, our observations have important clinical implications. The ability of Th2 biased infections to deviate the immune response to other antigens is well documented. Our model extends these findings in a novel direction by showing that enteric infection can elicit a Th2-biased immune response to a normally tolerogenic form of antigen. This suggests that helminth infection may pre-dispose for allergic phenomena and may affect the response to oral vaccines. In Specific Aim 2, we will therefore examine the ability of helminth infection to prime for an allergic response to a food antigen and explore the relevance of these findings to clinical food allergy. We will also examine the practical implications of mucosal parasitic infection for oral vaccination. If helminth infection biases for a Th2 response to oral vaccination, the success of oral vaccination campaigns in developing countries may hinge upon controlling the ability of these infections to alter responses to vaccine antigens. Characterization of the influence of mucosal infection on the response to oral vaccination using this murine model may suggest methods for maximizing the desired response to oral vaccines. | {
"pile_set_name": "NIH ExPorter"
} |
The aim of the research is to test the performance in animals of an innovative stentless chordally supported mitral valve designed to produce hemodynamics significantly superior to all existing devices, in vitro wear performance significantly exceeding current bioprosthesis, and treated so that calcification, in vivo, is inhibited and host endothelial covering occurs. The valve can be made quickly and cheaply. The testing will follow FDA Heart Valve Replacement Device Guidelines so as to complete the requirements for application for an IDE and an Export License. Once the IDE & Export License are granted, clinical trials following the Guidelines will start in the U.S.A., and overseas. It is anticipated that the testing will confirm the previous in vitro and in vivo results. | {
"pile_set_name": "NIH ExPorter"
} |
This project continues to explore the contribution of altered cholesterol me@abolism to the accelerated atherosclerosis of diabetes. We are studying a) the effect of diabetic control on cholesterol synthesis as determined by sterol balance and by the method of squalene kinetics; b) the effect of diabetes, ketosis and insulin on the rate-limiting enzyme in cholesterol synthesis HMG-CoA reductase; and c) the effect of diabetes on non-sterol pathways of mevalonic acid metabolism. The overall purpose of the study is to improve our understanding of how diabetes affects cholesterol metabolism, and how insulin treatment modifies these effects. | {
"pile_set_name": "NIH ExPorter"
} |
p53 tumor suppressor is a transcription factor and can be activated in response to various stress signals, including DNA damage, oncogene activation, and hypoxia. Upon activation, p53 exerts its tumor suppression by regulating a plethora of genes involved in the control of the cell cycle, apoptosis, DNA repair and other cellular processes. p21, a cyclin-dependent kinase inhibitor, is transcriptionally regulated by the p53 family to induce cell cycle arrest. p21 is also regulated post-transcriptionally upon DNA damage in a p53-dependent manner, but the mechanism is uncertain. During the last funding period, we identified several novel genes, which can be induced by p53 and DNA damage in a p53-dependent manner. Among these is the RNPC1 gene, which encodes at least two alternative spliced isoforms, RNPC1a and RNPC1b, both of which contain an intact RNA recognition motif. We found that over-expression of RNPC1a, but not RNPC1b, is capable of inducing cell cycle arrest in G1. We also found that while both isoforms directly bind to the 3'-untranslated region in p21 transcript, only RNPC1a is able to stabilize both the basal and stress-induced p21 transcripts. Conversely, RNPC1a knockdown decreases the basal level of p21 transcript. Thus, Specific Aim 1 is proposed to test the hypothesis that an mRNA decay pathway is regulated by RNPC1 to modulate p21 mRNA stability. The activity of p53 is primarily controlled by post-transcriptional mechanisms. While it is well known that modifications of p53, such as phosphorylation and acetylation, stabilize and activate p53, increasing evidence indicate that other post-transcriptional mechanisms play a major role in modulating p53 activity. For example, ribosomal protein L26 and nucleolin are found to bind to p53 transcript and positively and negatively regulate p53 translation, respectively. In addition, HuR is found to enhance p53 translation as well as p53 mRNA stability. Interestingly, HuR is also found to bind to p21 transcript and regulate p21 mRNA stability. As a RNA binding protein and a target of p53, we found that RNPC1 binds to p53 transcript and knockdown of total RNPC1 or RNPC1a increases the basal levels of p53 and further enhances p53 accumulation induced by DNA damage. Conversely, we found that over-expression of RNPC1a, but not RNPC1b, inhibits the basal levels of p53 and DNA damage-induced p53 accumulation. These data suggest that p53 is likely to be regulated by RNPC1. Furthermore, recent studies demonstrated that p53 translation is increased under stress conditions and our preliminary studies indicate that the efficacy of p53 translation is modulated by RNPC1 under both normal and stress conditions. Thus, Specific Aim 2 is proposed to test the hypothesis that RNPC1 regulates the cap-dependent translation machinery to modulate p53 translation under normal conditions and the cap-independent translation machinery to modulate p53 translation under stress conditions. [unreadable] [unreadable] PUBLIC HEALTH RELEVANCE: Regulation of p53 activity has always been at the center of p53 research as the majority of human cancers have a defective p53 or a faulty p53 pathway. The proposed study to analyze how RNPC1, as a p53 target gene, regulates p53 is highly significant in several areas. First, it will further our understanding of the p53 biology, which is needed in order to explore the p53 pathway for cancer diagnosis and treatment. Second, RNPC1 may be explored as a molecule target to activate p53 to sensitize tumor cells which contain wild-type p53, or to repress wild-type p53 to de-sensitize normal cells, to radiation- and chemo-therapies. Third, as a regulator of p53 translation, RNPC1 would regulate expression of mutant p53 in tumor cells. Since mutant p53 is an oncogene, RNPC1 may be explored to repress mutant p53 and thus, inhibit tumor growth and/or sensitize tumor cells to radiation- and chemo-therapies. [unreadable] Due to loss of p53 and other factors that are required for p21 expression, tumor cells are often dysfunctional in the cell cycle control, in which p21 is a key player. Restoration of proper cell cycle control in tumor cells is one of the many approaches that have been explored to suppress cell transformation and tumor progression and metastasis. We showed that p21 mRNA stability is regulated by RNPC1 under both normal and stress conditions. Thus, the proposed study to analyze how p21 expression is regulated by RNPC1 will further our understanding whether a cell cycle control mechanism can be restored in tumor cells. [unreadable] [unreadable] [unreadable] | {
"pile_set_name": "NIH ExPorter"
} |
Recently microarray technoloy has been implemented for measurements such as genome wide transcription patterns, and chromation immunoprecipitation analysis using genomic fragment arrays has been shown to be feasible for mammalian cells. Large scale gene knockdown experiments are also now achievable for analysis of gene function in cells from multizoates. Multipotential mesenchymal cells have been identified that can be propagated indefinitely amd differentiate within weeks into any one of a number of different cell lineages, in a manner controlled by the culture conditions. In addition to sequence specific DNA binding proteins, gene expression can be modified by the state of DNA methylation and the types and positions of modified residues in the core histones of nucleosomes. Further groups of proteins have been identified genetically and biochemically that function to stabilize gene activity or silence and interact with or participate in chromatin modification. The ability to rapidly undergo commitment to any of a number of cell lineages may be coupled to either limitations in the mechanisms for maintenance of gene activation or silencing or efficient functioning of mechanisms to reverse silenced and activated states of genes. Application of the new generation of genomic techniques could give fundamental insight into the mechanisms of multipotentiality and lineage choice and potential guide experimentation towards the development of regenerative medicine. | {
"pile_set_name": "NIH ExPorter"
} |
DESCRIPTION: The objective of this work is to advance the state-of-the- art in electrospray/ion trap mass spectrometry of biomolecules by improving the analytical figures of merit of the instrumentation as well as by developing new experiments to gain information on high mass biologically relevant ions. The quadrupole ion trap is a remarkably powerful tool for the manipulation and mass/charge analysis of biologically-derived ions. Furthermore, it is far less expensive than other forms of instrumentation capable of performing tandem mass spectrometric experiments. The latter characteristic makes the ion trap potentially accessible to a wide range of researchers in the biomedical field. This work seeks to achieve ultra-low level analysis/detection of high mass biomolecules (low attomole levels), without preconcentration, in common analytical scenarios such as on-line capillary electrophoresis and flow injection analysis. Particular emphasis will be placed on the use of non-destructive ion detection and re-measurement within the context of multi-stage mass spectrometry experiments. The ability to store ions will be used to develop new applications for ion/molecule reactions and to explore the chemistry associated with reactions of multiply-charged biopolymers with oppositely charged ions. The ion/molecule reaction applications include a technique to increase ion currents derived from multiply-charged biopolymers by several orders of magnitude via proton transfer reactions and an approach to dealing with electrospray of mixtures involving tandem mass spectrometry and proton transfer reactions. Ion/ion reaction studies will focus on both proton transfer and electron transfer with particular attention paid to the chemistry of the odd-electron species formed via electron transfer reactions. In all cases, emphasis will be placed on experiments that can provide new information or that can enhance the performance of the ion trap over current methodology. | {
"pile_set_name": "NIH ExPorter"
} |
We plan to look at latency in acute HIV infection. Using flow cytometry, we plan to purify different cell populations from whole blood and lymphnodes of patients with acute HIV infection. We will determine what percentage of different cell populations are infected and we will monitor how this changes with time. Using PCR, we will determine how soon latency is established in the course of acute infection. We will also determine how much of the drop in viremia after seroconversion is due to trapping of virions as immune complexes versus the elimination of a subpopulation of infected cells. We will also determine whether latency can be prevented by the early initiation of highly antiretroviral therapy. | {
"pile_set_name": "NIH ExPorter"
} |
The Malaria Genetics Section of the Laboratory of Parasitic Diseases conducts basic research on factors that govern the drug response, pathogenesis and transmission of malaria. The work incorporates strategies of linkage mapping, field population surveys, gene manipulation and gene product analysis with a view toward the discovery of fundamental biological information that will be of use in the development of new diagnostics, therapeutics and control measures against the disease. The research of the section largely focuses upon Plasmodium falciparum, the parasite that causes the most severe form of malaria and produces 2-3 million deaths each year from the disease. Current projects include investigations of 1) molecular mechanisms of drug resistance, particularly the resistance of malaria strains to such crucial anti-parasite drugs as chloroquine, quinine, and mefloquine; 2) gene transcription switches and DNA recombination events responsible for the antigenic variation and immune evasion of parasitized red blood cells; 3) epidemiology of hemoglobins C and S (sickle-cell) and their protection against severe malaria in African children; 4) a genetic defect in chromosome 12 that adversely affects the development of male gametocytes. - malaria; drug resistance; antigenic variation; hemoglobins; sickle-cell; chloroquine; quinine; mefloquine; genetics; genomics - Human Subjects & Human Subjects: Interview, Questionaires, or Surveys Only | {
"pile_set_name": "NIH ExPorter"
} |
In 2008, the Universidad Central del Caribe (UCC) revised its Institution's Research Strategic Plan, and in the present RCMI Renewal Research Application (2009-14) is requesting support for its concomitant implementation in compliance with the overall RCMI goal. The research plan responds to emerging needs by core facilities, research centers and individual projects which represent the scientific strength of our research enterprise. These activities will bring our research capabilities to another level of excellence by contributing to an increase in publications, presentations at scientific meetings, and funding available for research by over 20-50 percent. The new application proposes several main objectives: continuing to develop an Administrative Component consisting of a Leadership Core, a Scientific Research Development Unit and a Data Management and Statistic Research Support Unit; strengthening the proposed existing research centers (Retrovirus and Neuroscience) and create the Cancer Research Center; providing support to five research pilot projects in the area of Retrovirology, Neuroscience and Cancer; providing support and leadership to existing and new research Resources areas and core facilities that have proven to be indispensable for basic and clinical research enterprises; establishing an ongoing RCMI evaluation plan to assess the progress in these areas using objective criteria to verify research productivity. In order to fulfill the proposed main objectives, the organizational structure of the RCMI Program is divided into four components. The Administrative Component which is constituted by a Leadership Core, (evaluation and mentoring plan), the Scientific Resources Development Unit, and the Data Management and Statistical Support Unit. The Research Centers Component is structured to include the Retrovirus Research Center which consists of a Leadership Core and an HIV and Substance Abuse Laboratory Core; the Cancer Research Center which is provided with a Leadership Core; and the Neuroscience Research Center which possesses a Leadership Core, a Behavioral Testing Facility, and the Neuronal and Glial Culture Facility. The third component groups the five pilot projects. The General Research Cores Component comprises the fourth unit with the Optical Imaging Facility, the Protein and Nucleic Acid Core Facility, the Common Instrumentation Area and Services, and the Biomedical Proteomics Facility. The strategic plan is designed to make each center a cluster of investigators, each with a specific disease-oriented focus. The Cores Component is an ancillary unit designed to serve the other centers. | {
"pile_set_name": "NIH ExPorter"
} |
Pre-mRNA splicing is a fundamental process required for the expression of most metazoan genes. Defects in splicing lead to many human genetic diseases, and pre-mRNAs containing multiple introns and exons can be alternatively spliced in a cell type, cell cycle, or developmentally regulated manner by joining different pairs of 5' and 3' splice sites. Insights into the basic mechanisms of pre-mRNA splicing and splice site recognition are therefore fundamental to understanding regulated gene expression and human disease. The overall goal of this research proposal is to understand the mechanisms involved in splice-site recognition and pairing of pre-mRNAs. During the previous funding period, we have developed quantitative assays to provide new insights into the mechanisms of splice-site pairing. In the next phase of investigation, we propose to determine the ?molecular events that lock splice sites into a pairing position and to analyze how the combinatorial contribution of multiple splicing signals influence exon inclusion. Specifically, we will determine the biochemical steps that lead to splice-site pairing in A complex (Aim 1). We will test the hypothesis that ATP hydrolysis during A complex formation drives the irreversible juxtaposition of alternative splice sites or exons. In Aim 2 we will determine how the spliceosome executes commitment to splice-site pairing. We will use immuno-depletion and RNAi approaches to test the hypothesis that a subset of U2 snRNP components and associated proteins (CUS2/Tat-SF-1, Prp5, SF3a120, and UAP56) is necessary for irreversible splice-site pairing. Aim 3 describes a systematic and quantitative approach to determine how the probability of exon definition and inclusion is influenced by the combinatorial contributions of variable splice sites, enhancers, silencers, and the exon/intron architecture. We will test the hypothesis that measures of exon inclusion can be quantitated and used to improve the predictability of constitutive and alternative splicing within the human genome. These experiments are important because 1) the commitment to splice-site pairing constitutes arguably the most crucial step during the splicing reaction because it determines the splicing patterns of pre- mRNAs, and because 2) a quantitative framework of combinatorial exon recognition will elucidate mechanisms of splicing regulation and allow to predict the intrinsic pattern of splicing from sequence analysis. [unreadable] [unreadable] [unreadable] | {
"pile_set_name": "NIH ExPorter"
} |
The major objective of this project is to study the role of mitochondria in oxysterol-induced apoptosis of human lymphoblastic leukemia cells. Oxysterols represent a class of potent inducers of apoptosis in various cell types, particularly leukemic lymphoid cells. Oxysterols also act as powerful transcriptional regulators of genes involved in cholesterol biosynthesis, cell growth and apoptosis. Although the molecular basis of oxysterol-induced apoptosis of lymphoid cells remains unknown, some studies have suggested that oxysterol induction of apoptosis is mediated by reactive oxygen species generation. Results obtained in the previous funding cycle show that oxysterol treatment of CEM cells induces a decrease in ATP levels and loss of mitochondrial membrane potential. This project will test the hypothesis that ROS damage particularly to mitochondrial DNA plays a role in oxysterol-induced apoptosis of human leukemia cells. Our hypothesis predicts that oxidative damage to mitochondrial DNA can lead to mitochondrial dysfunction and induction of apoptosis. To test this hypothesis we will: 1) determine whether the formation and repair of oxidative damage to nuclear and mitochondrial DNA are higher in oxysterol sensitive human lymphoid cells than in oxysterol-resistant cells after exposure to oxysterols, 2) determine mitochondrial function and dysfunction in oxysterol-sensitive and resistant human lymphoid cells after exposure to oxysterols, and 3) determine the expression profile of genes involved in oxidative stress, DNA repair and apoptosis during oxysterol-induced apoptosis of lymphoid cells. This project will assist in laying the foundation for a research project directed towards the understanding of the role of mitochondria in the process of apoptosis. | {
"pile_set_name": "NIH ExPorter"
} |
The Biometric Research Branch (BRB) is the statistical component for scientific planning and monitoring of the national and international research program of the Division of Cancer Treatment. The branch provides statistical leadership for all extramural activities of the division and conducts collaborative research with the intramural components. The Biometric Research Branch performs statistical planning, monitoring and evaluation of all Division of Cancer Treatment supported therapeutic clinical trials. Primary statistical direction is provided by the branch for the conduct of selected national and international studies of therapeutic interventions, prognostic factors, pre-clinical screening and diagnostic imaging. The branch performs evaluations of therapeutic interventions based upon syntheses of results from multiple studies. The Biometric Research Branch conducts research on experimental designs, biometric methods and biomathematical approaches for the development and efficient evaluation of improved cancer treatments. | {
"pile_set_name": "NIH ExPorter"
} |
This research utilizes the Averaged Evoked Potential (AEP) technique to study human and animal brain function with the following objectives: to identify the neural structures whose activity is reflected in the human AEP and the neural pathways which participate in the activation of these structures; to analyze psychological phenomena which covary with alterations in the various components of the AEP; and to apply the knowledge of AEPs thus gained to clinical medicine, primarily neurology, for diagnosis and prognosis, especially prognosis for recovery of brain function, in various types of sensory neuropathology. During the period of this report we have completed or progressed on the following experiments: Topography of the averaged movement potential; Origin of early auditory evoked potentials in man: Effects of barbiturate anesthesia; Specific and nonspecific late components of the human SER; SERs recorded from the pial surface of the human brain; Studies in experimental epilepsy: 2. Effects of monomethylhydrazine (MMH) and other convulsant agents on somatic evoked potentials in the cat; Modification of EEG through biofeedback techniques; Evoked potentials in patients with epilepsy; Levels of processing in speech perception: Neurophysiological and information-processing analysis; AERs associated with temporal order judgments of dichotically presented sounds during linguistic and nonlinguistic processing; The relation between long-latency AERs and perceptual discrimination; An improved objective method for detection of absolute auditory threshold in evoked response audiometry; studies of comparative and evolutionary aspects of sleep; and sleep in the armadillo, Dasypus novemcinctus. | {
"pile_set_name": "NIH ExPorter"
} |
The Large Scale Antibody and T Cell Epitope Discovery Program supports the large scale discovery of novel B and T cell epitopes that utilize recent technological advances such as, but not limited to: computer-based epitope prediction algorithms;genome-wide scanning;structural genomic;high pressure liquid chromatography;mass spectrometry;phage-display libraries;and combinatorial synthetic peptide library screens. In addition, NIAID seeks to develop new or improved high throughput screening methods for epitope discovery, with a strong interest in antibody epitope screening methods. This program emphasizes epitope discovery for category A-C bioterrorism agents and emerging/re-emerging infectious diseases. | {
"pile_set_name": "NIH ExPorter"
} |
Chemotaxis is a fascinating biological response in which cells orient themselves and move up a chemical gradient. It is important in a variety of physiological and pathological processes including nerve growth, angiogenesis, wound healing, leukocyte trafficking, and carcinoma invasion. It is also essential for the survival of the social amoebae, Dictyostelium discoideum. During growth, these cells track down and phagocytose bacteria. When starved, they enter a differentiation program that allows the cells to survive harsh environmental conditions. They do so by chemotaxing toward secreted adenosine 3,5 cyclic monophosphate (cAMP) signals thereby forming aggregates which differentiate into spore and stalk cells. The essential role of chemotaxis in this eukaryote has provided an excellent model organism to study the biochemical and genetic basis of directed cell migration. Much like their mammalian counterparts, Dictyostelium cells use G protein-linked signaling pathways to respond to chemoattractants. Binding of chemoattractants to serpentine receptors leads to the dissociation of heterotrimeric G proteins into alpha- and beta/gamma-subunits, which activate a variety of effectors that go on to produce multiple responses. These include increases in Ca2+ influx, IP3, cAMP and cGMP. Concomitantly, the level of phosphorylation of myosins I and II and polymerized actin are markedly increased. Our research program is focused on understanding how these multiple G protein-coupled signaling events are translated into directed cell migration. We have shown that a variety of signaling events are spatially restricted during chemotaxis. In one instance, this has led us to discover a novel and unexpected mechanism used by Dictyostelium cells to relay and amplify chemotactic gradients. It has been observed that these cells align in a head to tail fashion, or stream, as they migrate in a gradient of cAMP. We have shown that the adenylyl cyclase ACA, which converts ATP into cAMP, is distributed in two distinct pools in polarized cells; one is restricted to the plasma membrane, and the other is localized on highly dynamic intracellular vesicles. These vesicles coalesce at the back of cells through mechanisms that depend on the actin and microtubule cytoskeleton, and require de novo protein synthesis. Further studies allowed us to propose that the asymmetric distribution of ACA provides a compartment from which cAMP is secreted to locally attract neighboring cells, thereby providing a unique mechanism to amplify chemical gradients. | {
"pile_set_name": "NIH ExPorter"
} |
PC4 is a human positive cofactor for RNA polymerase II to mediate activator-dependent transcription through the interactions with both activation domains and the basal transcription machinery. We have found that the function of PC4 is modulated by phosphorylation. Both protein-protein interaction and in vitro transcription assays demonstrate that only unphosphorylated PC4 is functionally active. Although in vitro phosphorylation experiments indicate that recombinant PC4 can be phosphorylated by casein kinase II and protein kinase C, the results from mutation analysis and determination of molecular masses of phosphorylated and dephosphorylated forms of natural PC4 by mass spectrometry suggest that PC4 is hyperphosphorylated mainly by casein kinase II in vivo in the first serine-rich region. These observations demonstrate a first example of transcriptional cofactor negatively regulated by CKII phosphorylation. A paper describing this work has been published in PNAS. Subsequent to the publication of the paper, we further refined our phorphorylation site mapping techniques utilizing the MALDI-Ion trap mass spectrometer. | {
"pile_set_name": "NIH ExPorter"
} |
The Division of Allergy and Infectious Diseases at the University of Washington has trained 115 physician-scientists through continuous funding provided by this training grant since its inception in 1976. More than 85% of these trainees remain in research positions. Many have achieved leadership positions such as two that became Deans of Medicine, one that became a Department Chair, 7 that became Division Heads, and 17 others with leadership positions. Trainees that have completed in the past 10 years have had remarkable early success in research careers, including >75% achieving research funding, >50% with K awards, and >90% publishing one or more papers related to their supported research. The continuing objective of the program is to provide post-doctoral training for physician-scientists committed to an academic career in infectious diseases, with particular emphasis on research training. Each trainee selects a mentor for detailed research training in one of six research tracks. All trainees complete a core curriculum consisting of required didactic, research and clinical activities. Many trainees elect to take course work at the University of Washington and an average of two a year seek a MPH or MSc award. The program generally requires three years of training, two of which are supported by this training grant. | {
"pile_set_name": "NIH ExPorter"
} |
Each year, over 30,000 patients suffering from hematological diseases such as leukemia, lymphoma, multiple myeloma, and severe aplastic anemia are treated with high-dose chemotherapy followed by hematopoietic stem cell (HSC) transplantations. The ability of HSCs to reconstitute all blood lineages post transplantation can be life-saving for these patients. However, their need is severely outweighed by their availability due to the difficulty in finding a suitable donor match coupled with their low expansion potential ex-vivo. A variety of culture conditions have been evaluated for in vitro expansion of HSCs but have yielded limited success due to an increase in differentiation, loss of stemness and promotion of cell cycle abnormalities in culture. Thus, this project aims to develop novel biomimetic biomaterial substrates and harbor the innate ability of co-existing niche cells to promote HSC expansion in vitro. Aim 1 will develop a biomaterial substrate to emulate the native bone marrow extracellular matrix environment and promote HSC expansion in vitro. Aim 2 will investigate if alternative sources of Nestin positive MSCs exist, which can be harnessed towards the expansion of HSCs in vitro. Finally, Aim 3 will determine the role of additional candidate niche cells in promoting HSC expansion in vitro. This proposal will characterize the molecular mechanisms behind HSC expansion due to the effects of the biomaterial or other candidate niche cells in culture with HSCs by analyzing cell surface receptors, cytoskeleton changes, soluble factors, genetic changes and associated intracellular pathways. Innovative methods for promoting HSC expansion could be used for life-saving transplantations to significantly impact the prognosis of patients with hematological malignancies. Additionally, this system can serve as a model for studying niche cell-cell interactions, cell-matrix interactions, drug screening and genetic diseases. These studies will significantly contribute to the knowledge base of the bone marrow niche microenvironment and potentially lead to the development of new patient therapies so they can live longer and more fulfilling lives. | {
"pile_set_name": "NIH ExPorter"
} |
Planning and Evaluation Core 7.1. Internal Advisory Committee Along with Drs. Hubbard and Rapkin, members of the Internal Advisory Core (IAC) provide major academic leadership and support for the Partnership. The LAC is organized in two major sections: translational research in cell biology and biomedical engineering and psychosocial research on health disparities. Both sections are involved in the development of transdisciplinary work to address disparities. Each IAC member has contributed significantly to our scientific and training activities. IAC members have responsibility for review of research and training programs, and have served as mentors and co-Pis on projects with junior faculty and trainees. Carol Brown. PhD Director, MSKCC Office of Diversity Initiatives. Dr. Brown is a member of the Psychosocial/Disparities IAC. As noted above, Dr. Brown is a gynecological surgeon with a focus on health disparities that affect minority women. Dr. Brown has helped to initiate work on women's health, including an Ros application submitted by Drs. Floyd and Ostroff to examine HPV vaccine uptake among inner-city families of adolescent daughters. Dr. Brown will work with the PCOP's cervical cancer initiative. She has also incorporated the Partnership's summer exposure program for high school students as a regular program of the Office of Diversity Initiatives. Nora Heaphy. MA. Coordinator of Service Learning Initiatives, Colin Powell Center for Policy Studies, CCNY. As a member of the Disparities IAC, Ms. Heaphy supports the Partnership's training and outreach activities. She has developed CCNY's program to train and support faculty in the development of service-learning curricula. Ms. Heaphy will also serve on our Education and Training Subcommittee. Jerard Hurwitz. PhD, Lab Head, Department of Molecular Biology, Vice-Chair, Sloan-Kettering Institute. Dr. Hurwitz is a member of our translational IAC. He is a molecular biologist whose research involves understanding DNA replication, its control and identification of proteins involved in this process. Dr. Hurwitz has been actively involved in helping CCNY investigators establish successful collaboration with MSKCC biologists and basic scientists, as well as promoting CCNY student placements in MSKCC labs. Maureen Killackev. MD. Deputy Physician-in-Chief, Head of the MSKCC Network. Dr. Killackey is a gynecological oncologist, and member of the Disparities IAC. As head of the MSKCC network, she is responsible for developing cancer treatment, support and outreach programs at satellite sites throughout the New York metropolitan area. Dr. Killackey is a member of the Board of the American Cancer Society's Eastern Division, and is a policy advisor to the NYS Department of Health Cancer Control Plan. Andrew Koff. PhD. Member, Department of Molecular Biology, Sloan-Kettering Institute. Dr. Koff is a member of the translational IAC. He is a cell and molecular biologist who studies cell cycle control in cancer and animal model systems. Dr. Koff has taken an active role in Partnership development, including assistance with animal containment issues at CCNY. He is serving as Dr. Hubbard's mentor, and with Dr. Hubbard, organized a highly effective symposium to bring together biological researchers from both institutions. He is also a member of our Executive Advisory Committtee. Robert Melara. PhD. Chair, Department of Psychology, CCNY. Since joining the Psychology Department as Chair in 2006, Dr. Robert Melara, has become actively involved with the Partnership's Internal Advisory Core, particularly in the area of health disparities. Dr. Melara has encouraged faculty to teach substantive and methodological courses related to cancer and health psychology, as well as service-based learning courses. He will continue to help the Partnership promote psychological research and methodological training related to cancer. Jamie Ostroff. PhD is Chief of the Behavioral Science Service and Associate Professor of Psychology at MSKCC. She is the Director of the Behavioral Research Methods Core Facility and the 732 supported Research Training Program in Psycho-Oncology at MSKCC. She has expertise in risk reduction, cancer screening, and family health communication. As a disparities IAC member, Dr. Ostroff leads the Partnership's efforts to provide on-going support and rigorous review for all projects. She is also MSKCC PI and Dr. Lubetkin's mentor on our pilot study of a navigation model to promote smoking cessation. Margaret Rosario. PhD is Associate Professor of Psychology and Director of the Health Psychology Concentration for the City University system. She is also a member of the Disparities IAC. Her work focuses on processes of identity development and their impact on mental and physical health. She has been instrumental in supporting and mentoring junior faculty in the social sciences, including leading the search committee that recruited Dr. Floyd. Mark Steinberg. PhD. Professor, Chemistry Department, CCNY. Dr. Steinberg is a member of the translational IAC. He is a cell and molecular biologist who studies viral transformation and factors controlling heavy metal induced carcinogenesis. Dr. Steinberg has been instrumental in building the cancer concentration within CCNY's Science Division. He is also involved in our efforts to undertake transdisciplinary research, including a Us6 pre-pilot with Dr. Manish Shah on gastric cancer disparities. John Tarbell. PhD Distinguished Professor and Chair, Department of Biomedical Engineering, CCNY. Dr. Tarbell is a member of the translational IAC. His interests include effects of fluid mechanical forces on endothelial permeability, cardiovascular fluid mechanics, and artificial heart and valve fluid mechanics. Overseeing rapid expansion, Dr. Tarbel has led the development of a major cancer research emphasis in his Department. Dr. Tarbell serves as principal investigator (with MSKCC Physician-in-Chief Wittes) of our collaborative P20 Biomedical engineering partnership, and is Dr. Wang's mentor on her pilot project. | {
"pile_set_name": "NIH ExPorter"
} |
We propose to begin a molecular analysis of the sensory properties of the fungus, Phycomyces blakesleeanus. Several mutations have been identified which block signal transfer at specific steps in the sensory transduction pathway. We plan to design a vector system for manipulating Phycomyces genes and to work out a procedure for introducing such vectors into Phycomyces by transformation or by microinjection. We will attempt to identify the sensory genes from a genomic library by their ability to restore function when introduced into host strains carrying the appropriate mutations in the sensory pathway. We can then hope to begin a detailed molecular analysis of these genes and their products to explore how their interactions lay down the sensory network. | {
"pile_set_name": "NIH ExPorter"
} |
Modulation of Oncogenic Signaling in Gliomas. A hallmark of human glioblastomas is that oncogenic signalings stimulated by overexpressed genes such as EGFR and EGFRvIII are aberrantly active, rendering these tumors highly malignant, and inherently resistant to combination therapies, resulting in an extremely poor prognosis of patients with malignant gliomas. A major barrier of this clinical challenge is the lack of a complete understanding of the mechanisms underlying the aggressiveness of these tumors and ineffectiveness of therapies for gliomas. The goal of this project is to define the role of ESDN, endothelial and smooth muscle cell-derived neuropilin-like protein (other names, DCBLD2 and CLCP1) in tumorigenesis and therapy-resistance of malignant human glioblastomas. ESDN is expressed at high levels in vascular injury, metastatic lung cancer and breast cancer. Our preliminary studies show that the ESDN gene is amplified in ~50% of clinical glioma specimens and preferentially up-regulated in classical and mesenchymal subtypes of glioblastomas that overexpress EGFR and EGFRvIII. Knockdown of ESDN in tumor xenografts significantly suppressed EGFRvIII-promoted glioma growth and invasion in the brain of animals. Mechanistically, EGFRvIII and EGF induce tyrosine phosphorylation (p-Y) of Y621 and Y750 of ESDN that activate TRAF6-Akt- and CrkII-Rac1- signaling and promote glioblastoma cell survival and invasion. Based on these observations, in this project, we propose a research plan directed at elucidating mechanisms by which ESDN augments EGFRvIII-stimulated glioma tumorigenesis and exploring therapeutic application of targeting ESDN to inhibit EGFRvIII-expressing brain gliomas. We will first investigate how ESDN affects EGFRvIII-stimulated glioma growth and invasion through dissociation of TRAF6 and binding of CrkII to specific tyrosine residues of ESDN that are phosphorylated by EGF and EGFRvIII stimulation. We will then determine clinical significance of the induced p-Y of ESDN with activation of oncogenic EGFR/EGFRvIII signaling pathways in primary glioma tumor specimens (Aims 1 and 2). Using short-term cultured primary glioblastoma (GBM) xenograft and an induced de novel brain glioma models, we will explore pre-clinical approaches to assess the impact of inhibition of ESDN on the efficacies of chemotherapeutic agents, cisplatin and temozolomide on EGFR- and EGFRvIII-expressing glioblastomas in the brain of animals (Aim 3). Therefore, this proposal reveals an unrecognized signaling path by which EGFR and EGFRvIII stimulate TRAF6-Akt- and CrkII-signaling through ESDN, thereby promoting glioma growth and invasion. Elucidation of this novel mechanism holds promise to a better understanding of the oncogenic signaling-promoted glioma tumorigenesis and to overcome the insidious glioma resistance to current therapies. This project also addresses an urgent challenge in neuro-oncology and offers an enormous potential for developing novel therapies that could prove effective, thus eliminating the major barrier in treating patients with malignant glioblastomas. | {
"pile_set_name": "NIH ExPorter"
} |
The objective of the administrative core is to facilitate and evaluate the operation of the Center. This includes supervising the operation ofthe six research projects and three additional cores, and facilitating communication and cooperation among Center personnel. | {
"pile_set_name": "NIH ExPorter"
} |
A fundamental question in cell biology is how the recruitment of proteins to cellular membranes is achieved and regulated. The long term objective of the proposed research is to characterize the structural and energetic basis for the binding of peripheral membrane proteins to phospholipid membranes and, in turn, to better understand the biophysical basis of membrane-mediated events in cells. The overall strategy is based on two distinct computational approaches: 1) the calculation of the physical interactions between proteins and membranes, and 2) bioinformatics tools for sequence analysis, structure comparison and structure prediction. By studying different membrane-interacting proteins in parallel, two hypotheses will be tested: 1) that two physical factors-electrostatics and hydrophobicity-are the major determinants of membrane binding, and 2) that these physical factors are manifested as patterns in sequence, structure and biophysical characteristics that can be used to predict membrane targeting potential. The first specific aim is to describe how nonspecific electrostatic and hydrophobic interactions mediate the wide range of membrane binding behaviors exhibited by secreted phospholipases A2. The second specific aim is to understand the role of electrostatic interactions in the calcium-dependent and independent membrane binding of C2 domains. The third specific aim is to develop structural models for phosphoinositides, an important class of signaling lipids. The fourth specific aim is to determine the energetic basis of both the specific and non-specific interactions of pleckstrin homology (PH) domains with membranes containing phosphoinositides. The computational results will be used in the design and interpretation of experiments through collaborations with experimental groups and will lead to rules that can be used to detect membrane targeting motifs in proteins. Secreted phospholipases A2 have been implicated in inflammation, and C2 and PH domain-containing proteins involved in phosphoinositide signaling have been implicated in oncogenesis; these proteins require membrane association for their function. Thus, a detailed understanding of the molecular mechanisms underlying the control of membrane association would facilitate the rational design of drugs that inhibit membrane binding. | {
"pile_set_name": "NIH ExPorter"
} |
The epidemiology of function, especially among the elderly and the oldest old is largely unstudied, particularly with representative samples of the elderly. The NHANES III survey with its commitment to sampling the elderly and to following the cohort, represents a unique opportunity for study. Physical function can be measured in a variety of ways: from self-report to observe functioning of actual daily activities. To permit efficient measurement of function in the elderly population, reliability of various techniques by expanding the continuum from a screening functional test to observed physical performance needs to be established. This proposal will develop instruments expanding this range for population studies in convenient samples of elderly living in the community in rural and urban areas and to do a one-year followup of the group. The emphasis of the sample is on those 65 and older and will also include a number of subjects over 75 years of age. The statistical properties of such measures will be established and hypotheses relating function to various care outcomes will be evaluated. | {
"pile_set_name": "NIH ExPorter"
} |
There are many diseases for which there are no vaccines and others for which the vaccines are not optimal or have significant side effects. The objectives of this project are to develop new vectors, characterize viral antigens, determine targets of humoral and cell mediated immunity, and use this information to develop candidate vaccines. Live recombinant viral vaccines, DNA vaccines and recombinant protein vaccines are being developed. Presently we are working on vaccinia virus-vectored vaccines for HIV. The vaccines are first tested in small animals and then in non-human primates. Phase 1 and 2 clinical trials with MVA-vectored vaccines developed in our laboratory are ongoing by collaborators. During FY2018, two phase 1 trials were completed and determined that the vaccines were safe and immunogenic. Animal studies revealed good boosting of MVA primed immunizations with gp140 and trimeric gp120 proteins and with virus-like particles. | {
"pile_set_name": "NIH ExPorter"
} |
Rabbits are the only or best animal model for several infectious diseases. The sequence of the rabbit genome is currently unfinished at 2x, but deeper 7x coverage started in 2007. A 2006 NHGRI recommendation that rabbit be sequenced more deeply, describes the NIAID allotype-defined rabbits as a valuable resource for future SNP discovery. They have polymorphisms of a variety of immune system genes including allelic allotypes of the VH, CH, and CL regions of antibody molecules. The colony also contained descendants of rabbits formerly at the Basel Institute for Immunology, including mutant and wild-type parental of VH1a2-deleted Alicia, CK1 splicing defective Basilea, and several VH-CH recombinant heavy chain types. These rabbits were made available to interested individuals, particularly to sites where breeding colonies could be established. A 4D relational database contains more than 45 years of breeding records and other information about animals in the colony. The mutant ali animals have a deletion of a key variable region gene in the immunoglobulin heavy chain locus that is present in related 2R1 wild type rabbits. The mutants may be more susceptibe to infectious disease because they have abnormal delayed development of humoral immunity. In part as the result of a White Paper I coauthored in 2005 proposing deep sequencing of the rabbit genome, 7x coverage was completed In July 2008 at Broad Institute MIT and Harvard. Assembly of the deep coverage is anticipated in early 2009. The sequences we have been able to find and use in the incomplete 2x trace archive, and in the 7x trace archive have already proven useful for various studies. As a result of my efforts, two websites are now available one maintained by the National Center for Biotechnology Information (NCBI) http://www.ncbi.nlm.nih.gov/projects/genome/guide/rabbit/ -- and one by NIAID on Rabbit in Immunology & Infectious Disease (http://www3.niaid.nih.gov/research/resources/ri/. The latter site offers a summary of an NIAID workshop on Rabbit Models of Human Infectious diseases held in 2005 that I helped the extramural NIAID DMID to program and chair. | {
"pile_set_name": "NIH ExPorter"
} |
Subsets and Splits