1992 — 1996 |
Wisniewski, Thomas M |
K08Activity Code Description: To provide the opportunity for promising medical scientists with demonstrated aptitude to develop into independent investigators, or for faculty members to pursue research aspects of categorical areas applicable to the awarding unit, and aid in filling the academic faculty gap in these shortage areas within health profession's institutions of the country. |
Diffuse Lewy Body Disease and Gelsolin
Diffuse Lewy body disease (DLBD) is emerging as a distinctive and common entity. It is characterized pathologically by numerous senile plaques of the Alzheimer's disease type and widespread cortical and subcortical Lewy bodies (LBs). Clinically there is a varying degree of dementia and parkinsonian symptoms. Some recent studies indicate that this neuropathology is the second commonest finding in cases of dementia. Lewy bodies are intraneuronal cytoplasmic inclusions first described in 1912; their pathogenesis remains unknown. Recently we have preliminary results showing that antibodies raised to purified amyloid extracted from familial amyloidosis, Finnish type (FAF) immunoreact with LBs. This immunoreactivity can only be absorbed by the purified amyloid and is unaffected by antigens such as ubiquitin. FAF amyloid has been found to be a degradation product of gelsolin, an actin modulating protein. The amino acid sequence shows heterogeneity at the N-terminus and at position 15 where asparagine substitutes for aspartic acid. At the DNA level a guanine to adenine transition corresponding to nucleotide 654 of the human gelsolin has been found in all FAF patients tested so far. We plan to: (1) Perform an immunohistochemical survey on DLBD, Alzheimer's disease (AD) and Parkinson's disease (PD) cases with the anti-FAF antibody, as well as numerous other antibodies to evaluate the pattern of immunoreactivity. In addition, the immunoreactivity of "eosinophilic bodies" reported in experimental models of PD will be studied, and compared to that noted in LBs. (2) Purify cortical LBs from a case of DLBD with biochemical methods, as well as using the anti-FAF antibodies for immunopurification. (3) Characterization of the composition of the purified LB preparation by the use of Western blotting and direct amino acid sequencing. (4) Eluting the protein bands which have been sequenced and using them to raise polyclonal antibodies. These will be assayed in LB containing material to verify that the material sequenced is contained in LBs. (5) Isolation of DNA from the brains of DLBD cases and familial PD to evaluate whether there is an abnormality in the gelsolin similar to that found in FAF or any other mutation. (6) Transmission studies with hamsters using the purified LB preparations. Together these studies will expand knowledge about LBs and the role gelsolin plays in their formation.
|
0.958 |
1997 |
Wisniewski, Thomas M |
S10Activity Code Description: To make available to institutions with a high concentration of NIH extramural research awards, research instruments which will be used on a shared basis. |
Fts 6000 Spectrometer Mainframe @ New York University School of Medicine
infrared spectrometry; biomedical equipment purchase;
|
0.958 |
1998 — 2002 |
Wisniewski, Thomas M |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Amyloid Beta Peptide and Their Binding Proteins @ New York University School of Medicine
DESCRIPTION (Adapted from Applicant's Abstract): The defining neuropathological lesions of AD are the deposition of amyloid beta (Abeta) in the form of amyloid fibrils in congophilic angiopathy and senile plaques, as well as the presence of neurofibrillary tangles. The Abeta in neuritic plaques is predominantly 1-42, while in vessels it is mainly Abeta1-40. Preamyloid lesions, the earliest types of Abeta deposits, are mainly Abeta17-42. Abeta peptides are also found in all biological fluids, with a main sequence of Abeta1-40. The latter is called soluble Abeta (sAbeta). Since Abeta peptides are produced throughout the body and can cross the blood brain barrier (BBB) in both directions, it remains unknown why amyloid deposition in AD occurs only in the brain. In this grant we will test the hypothesis that the clearance of brain sAbeta and its deposition is significantly affected by its binding proteins, such as apolipoprotein (apo)E. In the plasma sabeta is thought to be bound to apoJ, albumin or transthyretin. On the other hand, we have preliminary evidence that brain sAbeta in AD patients is partially bound to apoE, a protein that does not cross the BBB. Previously we have reported that in neuritic plaques amyoid Abeta is partially complexed to a carboxyl fragment of presenilin-1, a protein which is linked to the majority of early onset familial AD. In this grant we will determine how apoE and other Abeta peptide binding proteins interact with Abeta1-42, 1-40 and 17-42, to influence their passage across the BBB from the brain, as well as their conformation, aggregation state, toxicity in tissue culture and their ability to bind to senile plaques in vivo. We plan to: 1) Determine how much sAbeta from brain tissue is complexed to apoE and other proteins in normal controls versus AD and DS patients of differing ages and apoE isotopes. This will ascertain the importance of brain sAbeta-apoE complexes in AD pathogenesis. 2) Determine the influence of apoE and other sAbeta binding proteins on the conformation of Abeta1-42, 1-40 and 17-42 using FT-IR, circular dichromism and other methods. The influence of these binding proteins in tissue culture will be determined. 3) Determine if labeled Abeta1-40, 1-42 and 17-42 alone and with different binding proteins can cross the BBB either from ventricular or systemic injections in a transgenic model of AD, as well as in aged monkeys, with vessel amyloid and parenchymal Abeta deposits. We will also identify if any of these labeled peptides are deposited on the Abeta lesions, in vivo. The latter could be used to develop a diagnostic test for AD.
|
0.958 |
2002 — 2011 |
Wisniewski, Thomas M |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Detection and Clearance of Ad Amyloid Lesions @ New York University School of Medicine
DESCRIPTION (Adapted from applicant's abstract): The definitive diagnosis of Alzheimer's disease currently requires post-mortem examination. Our preliminary evidence shows that the Abeta peptide can be labeled with gadolinium (Gd) and be used following systemic injection for the detection of amyloid lesions in vivo in AD transgenic mouse models by magnetic resonance imaging (MRI) (using APPSW and APPsw/PS1 Tg mice). Gadolinium is currently widely used in clinical imaging as a contrast agent. A majority of parenchymal amyloid deposits can be detected using this method, providing potential approaches for both the early detection of amyloid lesions (when they may be most amenable to therapeutic intervention) and also monitoring the response to amyloid clearing therapy in vivo. Both these goals have become critically important since the recent demonstrations that immunization with human Abeta1-42 reduces Abeta deposition and prevents cognitive decline in Tg models of AD. However, this immunization method may be problematic in humans since we and others have shown that Abeta1-42 crosses the blood-brain barrier (BBB), where it is neurotoxic and can seed amyloid formation. In addition, the Freund's adjuvant used in these immunization experiments is too toxic for human use. Our preliminary evidence suggests that vaccination with non-fibrillar, non-toxic Abeta homologous peptides produces dramatic amyloid reductions in Tg AD models. In this application we plan to further develop the amyloid imaging methodology by altering the ligand to limit any potential toxicity and to increase BBB permeability, as well as using single chain and other anti- Abeta Gd labeled antibodies for imaging. We will also further test the efficacy of our non-toxic Abeta homologous peptides for vaccination in conjunction with alum based adjuvants (which are approved for human use), as well as testing passive immunization with anti- Abeta antibodies, which our preliminary results show can disaggregate pre-formed Abeta peptide fibrils. Importantly we will be able to subject immunized Tg mice to in vivo MRI in order to follow amyloid clearance. Vaccinated and imaged mice will be subject to behavioral testing to determine if cognitive deficits can be prevented by this approach. Our preliminary results indicate that APP/PS1 Tg aged mice have significant cognitive impairments versus controls. These studies will provide essential information before such approach can be safely used in humans.
|
0.958 |
2004 — 2013 |
Wisniewski, Thomas M |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Therapeutic Approaches For Prion Disease @ New York University School of Medicine
DESCRIPTION (provided by applicant): Prion diseases are infectious, conformational neurodegenerative disorders characterized by the structural modification of the prion protein, PrPC, into a pathological conformer, PrPSc. Currently there is no effective therapy for this group of diseases. The outbreak of bovine spongiform encephalopathy and the resulting emergence of a new human prion disease vCJD, highlight the public health threat from prion diseases. Although the original outbreak of vCJD is waning, there is the possibility of further outbreaks from current asymptomatic carriers of the disease. In the USA an ongoing threat from prion disease is from chronic wasting disease (CWD). High rates of infection among deer and elk populations have been report, with experimental data indicating that this disease is transmissible to primates. In the prior funding of this grant we reported on the first successful in vivo active and passive immunization approaches for prion diseases using wild-type animals. In the last funding cycle we also developed an in vitro tissue culture model of prion disease, and showed that it is a valuable tool to screen for therapeutically active anti-PrP antibodies. Our recent preliminary results indicate that using our novel mucosal immunization approach we are able to completely prevent prion disease among animals with a high anti-PrP titer. We also have shown that anti-PrP antibodies that are active in our tissue culture model of prion infection, can significantly delay prion infection and reduce severity of disease. In our planned studies we will further develop our active and passive immunization approaches, to bring them closer to veterinary and human clinical use for both prion infection prevention and potential treatment of symptomatic disease. The specific aims of this proposal are to : 1): Development of the optimal oral vaccination method in wild-type mice and transgenic mice expressing elk, sheep or human PrP; 2) Characterization of the immune response in successfully vaccinated animals and the development of monoclonal antibodies (Mabs) to mouse and human PrP from these mice. Mabs will be tested for therapeutic efficacy in tissue culture models of scrapie and human sporadic CJD prion infection and 3) Passive immunization studies using existing Mabs and antibodies generated from successfully vaccinated mice to determine the mechanisms responsible for therapeutic effects and to investigate if any will be effective in both human and scrapie prion infection in the pre- symptomatic incubation period and in symptomatic disease.
|
0.958 |
2005 — 2009 |
Wisniewski, Thomas M |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Amyloid Beta Peptide and Apolipoprotein E @ New York University School of Medicine
DESCRIPTION (provided by applicant): Inheritance of the apolipoprotein (apo) E4 allele is the major identified genetic risk factor for sporadic Alzheimer's disease (AD). We suggested in 1992 that apoE, especially its apoE4 isoform, can promote the conformational change of amyloid beta (Abeta) from soluble Abeta to toxic aggregates and amyloid plaques and coined the term "pathological chaperone" to describe this role. Many subsequent studies have been consistent with this hypothesis. In this proposal, based on our preliminary data, we plan to develop a novel, non-toxic therapeutic strategy for amyloid clearance in Alzheimer's disease, by blocking the apoE/Aa interaction. Based on the known binding site of apoE on Aa (Aa residues 12-28), we synthesized an end-protected, D-amino acid peptide Abeta12-28P, where the valine at residue 18 is substituted by proline. This modification renders the peptide non-toxic and non-fibrillogenic, without affecting binding to apoE. Abeta12-28P is blood-brain barrier permeable (clearance=65q20ul serum/g) and its serum half-life is 62q18 min. Our data shows this peptide reduces the Abeta/apoE toxicity and fibril formation in vitro. Transgenic APP/PS1 AD mice treated for one month with Abeta12-28P have a 63% reduced amyloid burden in the cortex (p=0.0048) and 62% in the hippocampus (p=0.0047) compared to controls. No toxicity was observed in the animals and no humoral response to Abeta was detected under our experimental conditions (so the treatment effect could not be due to antibody clearance of Abeta). Our preliminary data also indicates this treatment can reduce or remove existing amyloid lesions as documented by in vivo 2-photon imaging. Our Specific Aims are: 1) To evaluate the effect of blocking the apoE/Abeta interaction on amyloid burden (by in vivo imaging and histology), neuronal pathology and behavior in Tg animal models of Alzheimer's disease. 2) To identify the Aa binding site on apo E, so that further inhibitors of the apoE/Aa interaction can be designed based on the apoE sequence where Abeta binds and to develop peptidomimetics based on both the Aa and apoE sequences. 3) To determine the effects of blocking the Abeta/apoE interaction on other apoE functions as assessed by hippocampal neuronal spine integrity and receptor binding. These studies will test our central hypothesis that blocking the Aa/apoE interaction can serve as a novel, non-toxic therapeutic target for Alzheimer's disease.
|
0.958 |
2005 — 2021 |
Wisniewski, Thomas M |
P30Activity Code Description: To support shared resources and facilities for categorical research by a number of investigators from different disciplines who provide a multidisciplinary approach to a joint research effort or from the same discipline who focus on a common research problem. The core grant is integrated with the center's component projects or program projects, though funded independently from them. This support, by providing more accessible resources, is expected to assure a greater productivity than from the separate projects and program projects. |
Neuropathology Core @ New York University School of Medicine
The Neuropathology Core of this ADCC has the function to provide post-mortem diagnoses on patients and control subjects enrolled in the clinical core and on other well documented AD cases and controls, who are obtained via our satellite at the Sun Health Research Institute. Also part of this NP core and unchanged from the prior grant period, is a Morphometric Component performed at the Institute for Basic Research (IBR) on Staten Island. The Morphometric Component of this core is essential for the careful characterization of AD and control brain tissue in terms of the volume of different brain structures, number of neurons, glial cells, amyloid load and neurons with neurofibrillary change. This information can then be used for precise clinico-pathological correlation. The Specific Aims of this Neuropathology Core are: 1. To conduct thorough postmortem examinations on ADCC patients (N=15-20/yr from NYU) and an average of 55/yr from the Sun Health Research Institute (average post-mortem time 2.83 hr.). 2. To maintain a bank of unfixed frozen and fixed tissue from the ADCC control and patients with AD or other dementing neurodegenerative conditions;as well as to bank plasma and CSF from these patients. 3. To provide tissue from control brains, AD and other neurodegenerative conditions to AD researchers within and outside this ADCC in order to augment clinical and basic research on AD. 4. To conduct morphometric studies on AD to establish better clinical neuropathological correlation, in particular to document the progression of the earlier stages of AD pathology. 5. To conduct anatomic and pathologic correlation with in vivo and post mortem MR scans in collaboration with the Neuroimaging Core. 6. To collaborate in the research efforts of the other cores and projects of the ADCC, as well as providing advice and facilities to investigators within and outside the ADCC.
|
0.958 |
2005 — 2007 |
Wisniewski, Thomas M |
R03Activity Code Description: To provide research support specifically limited in time and amount for studies in categorical program areas. Small grants provide flexibility for initiating studies which are generally for preliminary short-term projects and are non-renewable. |
Mucosal Vaccination For Prion Disease @ New York University School of Medicine
DESCRIPTION (provided by applicant): This research will be done primarily in Uruguay at the University of Uruguay in collaboration with Drs. Jose A. Chabalgoity and Fernando Goni, as an extension of NIH grant #NS047433. Currently, there is not effective form of treatment for prion disease or other conformational disorders, such as Alzheimer's disease (AD). The pathogenesis of prion disease is related to a conformational change of the normal prion protein, PrPC, to a form with a high beta-sheet content, PrPSc. Recently vaccination has been shown to be an effective therapy in both AD and prion model mice by us and others. In the parent grant R01NS047433, we are focused on a variety of novel therapeutic approaches for prion disease, including vaccination in transgenic prion mouse models and metal chelation, along with imaging studies to correlate with a potential treatment effect. A significant problem with immunization for prion disease or other conformational disorders is toxicity. A human Phase II trail of active immunization for AD was terminated due to the development of toxicity in 6% of patients. However, patients without toxicity improved suggesting that the vaccination approach is effective. The toxicity of this vaccine has been linked to cell mediated immunity, while the efficacy of the vaccine approach is thought to be related to antibody mediated clearance of the disease associated protein. In this collaborative grant we plan to develop methods for inducing mucosal immunity in prion model mice. These studies are an extension of the work proposed in the parent grant. We have extensive new preliminary data showing the effectiveness of this approach, done in collaboration with the foreign scientists. Our vaccination approach will expression the prion protein in a Salmonella vaccine strain or coupling it to cholera toxin. These immunization approaches will primarily induce systemic and mucosal antibody responses. This immunization approach should be less likely to be associated with cell mediated toxicity. Induction of mucosal immunity may also be particularly effective for prion related disease, since the gastro-intestinal tract is thought to be the primary source of entry of the infectious agent. The proposed studies may lead to a mucosal prion vaccine that could be used for humans at risk to develop new variant Creutzfeldt-Jakob disease or animals at risk for prion diseases such as chronic wasting disease. These studies will build research capabilities in the foreign site by expanding their studies to include neurogenerative disorders and prion model animals.
|
0.958 |
2007 — 2008 |
Wisniewski, Thomas M |
R21Activity Code Description: To encourage the development of new research activities in categorical program areas. (Support generally is restricted in level of support and in time.) |
Immunization Approaches For Alzheimer's Disease @ New York University School of Medicine
[unreadable] DESCRIPTION (provided by applicant): This proposal seeks to develop a collaboration between the PI and Dr. Goni and Dr. Chabalgoity at the University of Uruguay. The Pi's laboratory is developing a novel immunization approach which is non-toxic for the treatment of Alzheimer's disease (AD) related amyloid in model mice, as well as vaccination to treat prion diseases. A major problem with vaccination approaches for these disorders in humans is potential toxicity. Both Dr. Goni and Chabalgoity are Immunologists, with Dr. Chabalgoity having a long experience with the development of various vaccines. The toxicity noted with the AD vaccine in humans has been linked to excessive cell mediated immunity, resulting in encephalitis in a about 6% of patients, whereas the beneficial amyloid clearing effects have been associated with humoral immunity. One way of inducing primarily a humoral, antibody mediated immune response is to induce mucosal immunity. We propose: 1. To develop mucosal active immunization for use in Alzheimer transgenic model mice using Salmonella vaccine strains, with the aim of inducing primarily a humoral immune response which will not be associated with toxicity. These will be used in conjunction with our Aft homologous peptides, which are non fibrillogenic and non-toxic. 2. Behavioral studies will be done and the amyloid burden will be determined in vaccinated and control AD model mice. The presence of any hemorrhages will be assessed. These histological measurements will be correlated with Ali peptide brain levels both in the soluble and insoluble fractions. 3. In the AD model animals the Th-1 versus Th-2 response will be monitored by the profile and magnitude of cytokine production as well as specific antibody titers in the Gl tract and systemically. In order to characterize the kind of humoral immune response generated, monoclonal antibodies will be produced using lymphocytes extracted from the Peyer's patches and spleens of mice with a positive outcome. This proposal will greatly enhance capacity building at the University of Uruguay with the transfer of AD transgenic model expertise and monoclonal antibody production capability. In addition it will produce preliminary data for the development of potentially non-toxic vaccination approaches for the treatment of AD. Lay Summary: AD is the most common cause of dementia. Vaccination is potentially an effective treatment, but is associated with significant toxicity in about 6% of patients. We propose studies to help develop safe, effective vaccines. [unreadable] [unreadable] [unreadable]
|
0.958 |
2010 — 2019 |
Wisniewski, Thomas M |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Therapeutic Targeting of Abnormal Conformation in Neurodegenerative Disease @ New York University School of Medicine
DESCRIPTION (provided by applicant): Many neurodegenerative diseases are characterized by the conformational change of self-proteins into amyloidogenic, pathological conformers, which share structural properties such as high [unreadable]-sheet content and resistance to degradation. Alzheimer's disease (AD) is the most common of the neurodegenerative protein conformational disorders, which include diffuse Lewy body disease (DLBD), Parkinson's disease (PD), prion diseases, and frontotemporal lobar degeneration (FTLD). The most toxic conformers are the oligomeric forms. None of the conformational diseases has an effective therapy;however, immunomodulation has shown great promise for both AD and prion diseases. Major problems with this approach include: the potential of toxicity from encephalitis (related to excessive cell mediated immunity), the immunological targeting of both the normal and abnormal A[unreadable], the resistance of vascular amyloid to clearance, as well as tau related pathology not being specifically addressed. The central hypothesis of this proposal is that each of these limitations can be overcome by specific targeting of abnormal oligomer conformation and development of novel methods to prevent oligomer mediated toxicity. Our novel active immunomodulation approach uses a polymerized British amyloidosis (pABri) related peptide in a predominantly [unreadable]-sheet, oligomeric form. We hypothesized that through "conformational mimicry" the polymerized ABri peptide could induce a conformation selective immune response that will recognize both A[unreadable] and conformationally abnormal tau. This hypothesis is supported by preliminary data in an APP/PS1 AD mouse model. Such an immunostimulatory approach should have a reduced risk of inducing auto-immune complications as it is more specific to a pathological conformer and the immunogen has no sequence homology to any known mammalian protein/peptide. We also present preliminary data that short term treatment with monoclonal 6D11, an anti-PrP antibody, reverses behavioral deficits in an AD model APP/PS1 Tg mice. This antibody blocks the binding of A[unreadable] oligomers to PrPC. We hypothesize that blocking the binding of A[unreadable] oligomers and PrPC is a novel therapeutic strategy for AD. These complementary approaches will aim to both increase clearance of A[unreadable] oligomers and specifically block their toxicity. PUBLIC HEALTH RELEVANCE: Many neurodegenerative diseases are characterized by the conformational change of self-proteins into amyloidogenic, pathological conformers, which share structural properties such as high [unreadable]-sheet content and resistance to degradation. Alzheimer's disease (AD) is the most common of the neurodegenerative protein conformational disorders, which include diffuse Lewy body disease (DLBD), Parkinson's disease (PD), prion diseases, and frontotemporal lobar degeneration (FTLD). The most toxic conformers are the oligomeric forms, which we plan to target by developing novel approaches to both increase their clearance and to reduce their toxicity.
|
0.958 |
2012 — 2016 |
Wisniewski, Thomas M |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Detection and Clearance of Ad Lesions @ New York University School of Medicine
DESCRIPTION (provided by applicant): The extracellular accumulations of amyloid ¿ (A¿) peptides as plaques and cerebral amyloid angiopathy (CAA), as well as intracellular neurofibrillary tangles (NFTs) are pathological hallmarks of Alzheimer's disease (AD). Treatments for AD available currently provide largely symptomatic relief with only minor effects on the course of the disease; hence, there is an urgent need for better therapeutic interventions. An intervention that is hoped to have a significant impact on disease progression in the near future is active or passive immunization with numerous past and on-going clinical trials. However, results so far indicate that despite successfully reducing plaque amyloid, the clinical benefits to patients are very limited. Major difficulties identified with the current approaches is that they specifically only target plaque amyloid, while having limited effect on CAA or may increase CAA and associated microhemorrhages and also limited or no effect on tau related pathology. The central hypothesis we plan to test in our proposal is that it is essential to develo and test therapeutic interventions which are effective against both amyloid plaques and CAA, as well as reducing tau related pathology. In this proposal we will further develop and test two approaches which we have already shown to be able to clear plaque amyloid, in the prior funded period of this grant. We hypothesize that these therapeutic approaches can also ameliorate both CAA and tau related pathology. We have previously shown that stimulation of innate immunity with the TLR9 agonist CpG ODN administration in Tg2576 AD model mice led to a remarkable reduction of the plaque amyloid burden which was associated with significant cognitive improvement, in the absence of any toxicity. We have preliminary data that TLR9 stimulation is also effective at reducing tau related pathology in 3xTg mice with both plaque and tau pathology. We have also shown that modulating the binding between A¿ and apolipoprotein (apo) E can diminish amyloid plaques using A¿12-28P (an inhibitor of A¿ and apoE binding) and dramatically reduce CAA in a vascular amyloid mouse model TgSwDI. In this proposal we will further test these two approaches in models of extensive CAA and in tau AD models for efficacy and safety. We will also test whether the treatment effect on the reduction of the CAA burden can be followed longitudinally in vivo using USPIO amyloid binding particles for mMRI on a subset of Tg mice in aims 1 and 2, using a modification of methods we have recently published. We believe our planned studies of these innovative methods to clear and detect AD pathology will have a significant impact on the field.
|
0.958 |
2015 — 2019 |
Wisniewski, Thomas M |
P30Activity Code Description: To support shared resources and facilities for categorical research by a number of investigators from different disciplines who provide a multidisciplinary approach to a joint research effort or from the same discipline who focus on a common research problem. The core grant is integrated with the center's component projects or program projects, though funded independently from them. This support, by providing more accessible resources, is expected to assure a greater productivity than from the separate projects and program projects. |
Core D. Neuropathology Core (With Ibr Subcontract) @ New York University School of Medicine
PROJECT SUMMARY/ABSTRACT NEUROPATHOLOGY CORE The Neuropathology (NP) Core provides for diagnostic neuropathological evaluations on patients and control subjects enrolled in the clinical core and on other well-clinically documented patients with neurodegenerative disease. The Core aims to gain enhanced understanding of the pathogenesis of AD, develop better biomarkers and discovery novel effective, therapeutic approaches. The protocols used by the NP core will be state of the art and consistent with 21st century brain banking procedures, which is essential to meet the increasingly sophisticated needs of the AD research community. Emphasis will be placed on delineating the presence of overlapping neurodegenerative syndromes such as vascular dementia, Lewy Body Disease, frontotemporal degeneration and prion disease, in addition to AD related pathology. In the current period of funding we performed over 130 autopsies, published over 65 peer reviewed manuscripts and provided tissue to over 50 investigators. In this renewal we will place emphasis on the center theme of this ADCC, which is elucidating the clinic-pathological substrates that underlie the transitions from normal aging to preclinical AD, preclinical AD to MCI, and MCI to the earliest stages of dementia. The overall objectives of the NP core of the NYU ADCC are to provide staff, technical resources, laboratory facilities and expertise for the investigation of structural and molecular properties of the brains of patients with AD and related dementia, as well as of normal controls. The aims of the NP core are: Aim 1: To perform rapid autopsies and obtain brains from ADC participants, using a standardized, state of the art protocol. Aim 2: Performed standardized state of the art neuropathological analysis and diagnosis of cases from the ADCC clinical core and affiliated cases, with a careful analysis of concomitant pathology. Aim 3: To maintain a state of the art brain tissue repository of unfixed frozen and fixed tissue from Aim 4: To conduct morphometric studies to establish better clinical neuropathological correlations for transitions of normal aging to preclinical AD, preclinical AD to MCI and MCI to early dementia. Aim 5: To foster training of investigators and residents on the utilization of neuropathological techniques, train neuropathologists and enhancing the use of Core tissue/resources for the identification of novel biomarkers and therapeutic approaches for neurodegenerative diseases.
|
0.958 |
2015 — 2019 |
Wisniewski, Thomas M |
P30Activity Code Description: To support shared resources and facilities for categorical research by a number of investigators from different disciplines who provide a multidisciplinary approach to a joint research effort or from the same discipline who focus on a common research problem. The core grant is integrated with the center's component projects or program projects, though funded independently from them. This support, by providing more accessible resources, is expected to assure a greater productivity than from the separate projects and program projects. |
Alzheimer's Disease Core Center @ New York University School of Medicine
DESCRIPTION (provided by applicant): The Alzheimer's Disease Center (ADC) at New York University Langone Medical Center (NYULMC) is an Alzheimer's Disease Core Center (ADCC) that provides outstanding infrastructure, environment and core resources promoting cutting-edge research by bringing together clinical, translational, basic and psychosocial investigators to study the etiology, pathogenesis, diagnosis, treatment and prevention of Alzheimer's disease and related disorders (collectively referred to as AD). Now entering its 25th year, the NYU ADC continues to provide a mechanism for fostering and coordinating the interdisciplinary cooperation of a group of established investigators conducting programs of research on AD while providing a supportive milieu for early-stage investigators. The ADC supports innovative research by maintaining an integrated array of core facilities, including Administrative, Clinical, Neuropathology, Data Management and Statistical, Outreach, Recruitment and Education, and Neuroimaging and Psychosocial Cores. The principal goal of the ADC is to enhance the performance of innovative AD research, including research that may lead to potential disease-modifying therapies. Along with the overall NIA-ADC program, the NYU ADC has moved into a new era, capitalizing on the extraordinary opportunities presented by leveraging the strengths of the network of centers to provide large numbers of samples and standardized clinical, cognitive and biomarker data from well-characterized participants, as well as a large pool of potential participants for future AD-related research. At the same time, we place a strong emphasis on the unique contributions and new directions of the NYU ADC. Additionally, emphasis is placed on possibilities for utilizing the resources within our ADC and across the ADCs to advance and augment drug discovery and drug development for novel therapeutics for AD. Since its inception in 1990, the NYU ADC has facilitated cutting edge research defining normal aging and the transition to the earliest detectable stages of mild cognitive impairment (MCI) and AD. This longstanding research direction will continue to be our main scientific theme, with a particular focus over the past 15 years on better understanding the preclinical stages that precede MCI and mild AD. Thus, we will continue our scientific focus on elucidating the transition from normal aging to the earliest, preclinical stages of AD. Our collective research efforts investigating the clinical-pathological changes associated with cognitive impairment and the role of co-morbid pathologies and chronic conditions position us well to develop, test and validate novel strategies to address symptomatic, disease-modifying, and preventive therapeutic approaches being developed at our ADC and other ADCs.
|
0.958 |
2016 — 2019 |
Wisniewski, Thomas M |
P30Activity Code Description: To support shared resources and facilities for categorical research by a number of investigators from different disciplines who provide a multidisciplinary approach to a joint research effort or from the same discipline who focus on a common research problem. The core grant is integrated with the center's component projects or program projects, though funded independently from them. This support, by providing more accessible resources, is expected to assure a greater productivity than from the separate projects and program projects. |
Core a. Administrative Core @ New York University School of Medicine
PROJECT SUMMARY/ABSTRACT ADMINISTRATIVE CORE The Administrative Core provides overall administrative supervision and coordination of the ADC to optimize the scientific productivity of the affiliated investigators and ensure that the basic objectives of the ADC are achieved. The general goal of the Core is to provide overall intellectual leadership and organizational cohesion to ensure an environment conducive to the conduct of productive research on brain aging and Alzheimer s disease (AD), focusing on elucidating the transition from normal aging to preclinical, mild cognitive impairment and early dementia stages of AD. The Core will continue to coordinate and facilitate achieving ADC goals and interact closely with each ADC Core and affiliated research project. It will set the overall direction of the Center and ensure optimal utilization of Center resources in support of ADC scientific objectives. This will be accomplished by continuing to provide the functions developed over the past twenty-four years in order to achieve the Core s specific aims. These include coordinating and integrating ADC components and activities; providing direction for future planning and optimal utilization of resources; supporting and advising the ADC Director in the oversight of the Center activities; interacting with the scientific and lay communities to develop relevant goals for the Center; organizing, maintaining, operating and coordinating meetings of Center advisory panels, including an External Advisory Committee to annually evaluate the programs and research progress of the ADC, an Executive Committee (composed of core leaders and the administrator) to assist the Director in making the scientific and administrative decisions, and a Pilot Review Committee to assist in annual selection of pilot projects; coordinating pilot projects; interacting with other Centers, the National Alzheimer s Coordinating Center (NACC) and other researchers; providing timely and routine transmissions of ADC data to NACC; ensuring interaction and involvement with other research programs at NYU; providing, implementing and supervising the resource sharing plan for the ADC as a whole, and coordinating with NIA on media coverage of the latest research findings from the ADC. In summary, the Administrative Core of the ADC is an essential component that sets the overall direction of the Center, coordinates its activities and ensures optimal utilization of Center resources. The management efforts of the Administrative Core are critical to the success of the ADC in achieving its specific aims.
|
0.958 |
2017 |
Haines, Jonathan L (co-PI) [⬀] Safar, Jiri G. Wisniewski, Thomas M |
RF1Activity Code Description: To support a discrete, specific, circumscribed project to be performed by the named investigator(s) in an area representing specific interest and competencies based on the mission of the agency, using standard peer review criteria. This is the multi-year funded equivalent of the R01 but can be used also for multi-year funding of other research project grants such as R03, R21 as appropriate. |
Characterization of Rapidly Progressive Alzheimer's Disease @ New York University School of Medicine
ABSTRACT The most frequent neurodegenerative disease is late-onset Alzheimer's disease (AD). We have recently reported a novel subgroup of patients who have a particularly malignant form of rapidly progressive late-onset Alzheimer's disease (rpAD) with atypical clinical symptoms, a low frequency of the e4 allele of APOE gene, unique structural characteristics of beta amyloid and an amyloid proteome that is distinct from typical slowly progressive Alzheimer's disease (spAD). Based on our preliminary data, we hypothesize that the rapid rates of cognitive decline and variable spectrum of symptoms in rpAD arise from the interplay between differently structured amyloid beta and tau proteins, triggering divergent pathogenetic cascades on a distinct genetic background. Accordingly, this proposal will focus on the integrated investigation of (i) conformational structural characteristics of beta amyloid and tau proteins with novel biophysical tools, (ii) proteomic profiling of amyloid plaques, neurofibrillary tangles, astrocytes and neurons of rpAD versus spAD, and (iii) genetic determinants linked to rpAD. The ultimate goal of these studies is to advance our understanding of the molecular mechanims governing the propagation of toxic beta amyloid and tau protein aggregates in the brain and the impact of their conformations upon the AD phenotype in the context of specific risk genes. This insight is critical for efforts to characterize key factors responsible for the very rapid rate of cognitive decline in this subtype of AD and ultimately to novel therapeutic strategies to slow AD progression.
|
0.958 |
2017 — 2021 |
Scharfman, Helen E (co-PI) [⬀] Wisniewski, Thomas M |
T32Activity Code Description: To enable institutions to make National Research Service Awards to individuals selected by them for predoctoral and postdoctoral research training in specified shortage areas. |
Postdoctoral Research Training in Neurodegenerative Disorders and the Aging Brain @ New York University School of Medicine
Project Summary This training grant application proposes the centralization of training postdoctoral candidates interested in a career in brain aging and neurodegenerative disease at NYU Langone Medical Center (NYULMC). There is a critical need to train the next generation of MD and PhD researchers to investigate causes of disorders of brain aging, develop new therapeutics and interventions, and translate these findings from bench to bedside as well as from bedside to community. This application is designed to select four outstanding postdoctoral trainees from a well qualified pool of MD and PhD applicants, develop and implement a comprehensive curriculum and provide 2-years of high quality research training in basic, translational, clinical, and psychosocial research. This Training Program, centered in the Center for Cognitive Neurology (CCN), a school-wide, extradepartmental, multidisciplinary program designed to foster collaborative, translational research. The CCN will serve as the unifying umbrella under which this program will be implemented, sustained and financially supported while providing the infrastructure to guarantee success and supplying the trainees a ?sense of community? and a home-base within NYULMC. The future demand for highly skilled investigators with a proven track-record examining brain aging and neurodegenerative diseases will be substantial as aging-related neurodegenerative disorders continue to increase with an aging population. The training of the next generation of research scientists and research clinicians with expertise in these diseases is and will remain a public health priority in the United States for many years to come. The Institute on Medicine recognized the dearth of junior investigators and clinicians with expertise in geriatrics and gerontology. To date, these calls for increased entrance into aging investigation have been left unanswered. Thus, a strong and compelling rationale exists to consolidate and expand the research-training program for brain aging and neurodegenerative disorders through the support of a T32 training program. To do this we propose the following Specific Aims: 1. Develop research training opportunities for postdoctoral candidates in the area of brain aging and neurodegenerative disease; 2. Educate trainees to fundamentals of brain aging and neurodegenerative disease, rigorous research methodology, clinical approaches, statistical analyses, and the principles of responsible conduct of research.; 3. Provide an interactive, collaborative environment fostering personal and professional growth for clinical, psychosocial, translational and basic science trainees.; and 4. Enhance trainees' requisite skills to gain external funding in the form of K- and R- awards.
|
0.958 |
2018 |
Busciglio, Jorge A Cuello, A Claudio Fortea, Juan Wisniewski, Thomas M |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
The Role of Inflammation and Ngf Dysfunction in the Evolution of Alzheimerdisease Pathology in Down Syndrome @ University of California-Irvine
PROJECT SUMMARY/ABSTRACT Individuals at risk of Alzheimer?s disease (AD) undergo a long, asymptomatic phase of disease progression that is characterized by the gradual accumulation of pathology in the absence of apparent cognitive deficit. Down syndrome (DS) patients represent a population at high risk of AD with complete penetrance of AD pathology in the majority of DS subjects, making DS an outstanding natural genetic model for the study of neuropathological mechanisms of AD and for identifying potential AD biomarkers. Given the current limitations in reliable early diagnostic tools, the discovery of biomarkers signalling the evolution of Alzheimer?s disease pathology in individuals at risk, including those with DS, is of utmost clinical relevance. The overall goal of the current proposal is to investigate novel, uncharacterized biomarkers at the earliest preclinical stages of AD in DS. To achieve this goal, a collaborative effort will integrate data from post-mortem brain studies, primary human cell cultures studies, and biological fluids (plasma and CSF) studies in a large well-characterized cohort of DS and matched control subjects. The central hypothesis of the proposal is that the early accumulation of intracellular amyloid beta peptide (A?) in DS brains will induce CNS inflammation accompanied by NGF metabolic dysfunction prior to extracellular amyloid plaque deposition. Furthermore, the CNS compromise of NGF metabolism should be detected in DS body fluids at ?incipient? stages of the AD pathology, before the presentation of overt dementia. To test the stated hypothesis, three Specific Aims will be accomplished: Aim 1: To explore the occurrence of a CNS pro-inflammatory process and NGF dysfunction throughout the lifespan in DS. Using post-mortem DS brains at different ages, the appearance of inflammation and NGF dysfunction in DS will be investigated to temporally reconstruct the evolution of AD pathogenesis. Aim 2: To investigate molecular mechanisms that link early AD pathology in DS with neuroinflammation and NGF metabolic dysfunction using fetal human primary cortical cultures. These studies will complement Aims 1 and 3 and will elucidate molecular pathways underlying early AD pathogenesis. Aim 3: To analyze the expression of A?, tau, NGF-related and inflammatory markers in matched plasma and CSF samples from DS subjects across the lifespan. These studies will test whether markers investigated in Aims- 1 and -2 are reflected in matched plasma and CSF samples from DS versus control subjects. This multi-PI multidisciplinary proposal will reveal fundamental molecular pathobiological mechanisms for AD in DS, it will identify biomarkers, and it will assist in the prediction of the onset and trajectory of dementia. In addition, the planned studies will likely lead to the identification of novel therapeutic targets in both DS individuals and sporadic AD populations.
|
0.911 |
2020 — 2021 |
Wisniewski, Thomas M |
P01Activity Code Description: For the support of a broadly based, multidisciplinary, often long-term research program which has a specific major objective or a basic theme. A program project generally involves the organized efforts of relatively large groups, members of which are conducting research projects designed to elucidate the various aspects or components of this objective. Each research project is usually under the leadership of an established investigator. The grant can provide support for certain basic resources used by these groups in the program, including clinical components, the sharing of which facilitates the total research effort. A program project is directed toward a range of problems having a central research focus, in contrast to the usually narrower thrust of the traditional research project. Each project supported through this mechanism should contribute or be directly related to the common theme of the total research effort. These scientifically meritorious projects should demonstrate an essential element of unity and interdependence, i.e., a system of research activities and projects directed toward a well-defined research program goal. P30Activity Code Description: To support shared resources and facilities for categorical research by a number of investigators from different disciplines who provide a multidisciplinary approach to a joint research effort or from the same discipline who focus on a common research problem. The core grant is integrated with the center's component projects or program projects, though funded independently from them. This support, by providing more accessible resources, is expected to assure a greater productivity than from the separate projects and program projects. |
Administrative Core @ New York University School of Medicine
CORE A- SUMMARY/ABSTRACT Our ultimate goal of this program grant is to gain a better understanding of the heterogeneous pathogenesis of AD and how it is influenced by apolipoprotein (apo) E isotypes, using human tissue, with correlations to findings in various AD models, using proteomic and other methods which we have recently developed. In particular we are focusing on the differential role apolipoprotein E (apoE) isoforms play in: 1) AD plaque and vessel amyloid development (Project 1); 2) therapeutic approaches that target the A?/apoE interaction (Project 2); 3) responses to therapeutic immunomodulation targeting abnormal conformation (Project 3). The Administrative Core (Core A) is charged with ensuring the success of this research proposal. The Aims are: Aim 1. Provide administrative structure and fiscal oversight for the program project grant. Aim 2. Organize regular meetings of the Executive Committee (EC) composed of Project and Core Leaders and other key personnel to assist in scientific administration and to facilitate integration and continuing progress on research aims. Aim 3. Coordinate with the bioinformatics/statistical/data management Core D to ensure optimal data utilization and scientific rigor for the Program Project. Aim 4. Interact closely with the External Advisory Committee and Internal Advisory Board, and conduct regular meetings of both. The External Advisory Committee and Internal Advisory Board will provide scientific advice and guidance for all research projects. Aim 5. Report progress to NIH and ensure compliance with the NIH Public Access policy. Aim 6. Promote education on AD and related disorders, and facilitate P01 scientists? participation in education to researchers, clinicians and patients/caregivers.
|
0.958 |
2020 — 2021 |
Wisniewski, Thomas M |
P01Activity Code Description: For the support of a broadly based, multidisciplinary, often long-term research program which has a specific major objective or a basic theme. A program project generally involves the organized efforts of relatively large groups, members of which are conducting research projects designed to elucidate the various aspects or components of this objective. Each research project is usually under the leadership of an established investigator. The grant can provide support for certain basic resources used by these groups in the program, including clinical components, the sharing of which facilitates the total research effort. A program project is directed toward a range of problems having a central research focus, in contrast to the usually narrower thrust of the traditional research project. Each project supported through this mechanism should contribute or be directly related to the common theme of the total research effort. These scientifically meritorious projects should demonstrate an essential element of unity and interdependence, i.e., a system of research activities and projects directed toward a well-defined research program goal. |
Blocking the Binding of a? and Apoe as a Novel Therapeutic Approach For Ad @ New York University School of Medicine
PROJECT 2- SUMMARY/ABSTRACT The pathological accumulation of A? peptides as toxic oligomers, amyloid plaques and cerebral amyloid angiopathy (CAA), either from increased production of A? peptides or from their inadequate clearance, is critical in the pathogenesis of Alzheimer?s disease (AD). The apolipoprotein E4 (apoE4) allele, a major genetic risk factor for late-onset AD, was strongly associated with increased amyloid plaque and vascular amyloid pathology. We have shown that using A?12-28P peptides to block the apoE/A? interaction constitutes a novel treatment for AD by reducing brain parenchymal and vascular amyloid burden as well as tau -related pathology in multiple AD transgenic (Tg) lines. We also showed that A?12-28P penetrates the blood-brain-barrier (BBB). In our preliminary studies, we designed a peptoid library derived from the A?12 -28P sequence to screen for apoE/A? binding inhibitors with higher efficacy and safety compared to A?12-28P. Cyclic peptoids typically have better cell permeability compared to the linear peptides of the same or similar sequence. Indeed, our lead peptoid CPO_A?17-21P is cyclic, has a Ki of 1.02 nM against the binding of apoE4 and A?, and has therapeutic efficacy in an APP/PS1 AD Tg model. Our preliminary experiments clearly show it is highly effective at reducing the amyloid burden at a dosage 7.5-fold lower than that used with the parent A?12-28P. CPO_A?17-21P is therefore an outstanding starting point for further biochemical and medicinal chemistry development of both novel peptoid and drug-like, small molecules. We propose testing our lead small, BBB- penetrant, peptoid molecule and analogous drug-like, small molecules in vivo, hypothesizing that these will reduce both neuronal and synaptic toxicity by inhibiting the apoE4/A? interaction. We will investigate how these treatments affect the amyloid proteome, and correlate changes to findings in human tissue ( Project 1) and the proteome in the same AD Tg models after immunotherapy (Project 3). We hypothesize that the treated amyloid proteome in apoE4 mice will convert to a more apoE3-like proteome. Aim 1: Design non-toxic, pharmacokinetically favorable peptoid and drug-like, small molecule antagonists of the apoE/A? interaction, and characterize their effects in vitro. Aim 2: Te st the lead peptoid and analogous drug -like, small molecules in vivo using 3xTg mice, APP/PS1 and TgSwDI mice crossed onto human knock-in (KI) apoE2, E3 or E4, or apoE knock-out (KO) backgrounds. Aim 3: Compare the amyloid plaque and vessel proteomes in peptoid and drug-like, small molecule tre ated Tg and control mice on KI apoE2, E3 or E4, or apoE KO backgrounds .
|
0.958 |
2020 — 2021 |
Wisniewski, Thomas M |
P30Activity Code Description: To support shared resources and facilities for categorical research by a number of investigators from different disciplines who provide a multidisciplinary approach to a joint research effort or from the same discipline who focus on a common research problem. The core grant is integrated with the center's component projects or program projects, though funded independently from them. This support, by providing more accessible resources, is expected to assure a greater productivity than from the separate projects and program projects. |
Biomarker Core @ New York University School of Medicine
ABSTRACT ? BIOMARKER CORE The primary objective of the newly configured Biomarker Core (BC) is to collect, bank, and distribute biofluids in order to generate biomarker datasets that will address disease heterogeneity and clinical transitions to subjective cognitive decline (SCD), mild cognitive impairment (MCI) and early stages of Alzheimer's disease (AD), with the long term goal of developing novel interventions that will delay or prevent these transitions. During its more than 25 years of continued funding, the NYU ADRC has been in the forefront of improving diagnostic tools and defining preclinical and prodromal stages of AD. In this renewal period, the dedicated BC will be reconfigured to serve as a conceptual and technical core for established investigators and the next generation of students and neuroscientists; and, to foster the use of plasma, serum and cerebrospinal fluid (CSF), as well as stool samples for microbiome analyses, to understand biological mechanisms of disease progression and heterogeneity. We propose the following Aims. In Aim 1, we will coordinate intake, processing and storage of biofluids from Clinical Core (CC) participants in association with the Data Management & Statistics (DMS) Core. In Aim 2, we will perform state-of-the-art analysis of known blood and CSF biomarkers for the diagnosis, prognosis, and prediction of AD. Assays will include, but will not be limited to, A?40/42/38, phosphorylated/total tau and neurofilament light-chain, all within the new amyloid/tau/neurodegeneration (ATN) framework, in addition to markers of cerebrovascular disease, inflammation, glial activation, as well as biological variables such as age, sex, ApoE4, and sleep (among others), which will be key to increase our understanding of disease heterogeneity. In Aim 3, we will use Simoa technology to develop novel ultrasensitive biomarker assays, guided in part by proteomic findings by the Neuropathology (NP) Core and other emerging data from the ADRC network. Aim 4 is designed to manage samples, perform quality control and analyses as well as link sample information obtained through the DMS, CC, Neuroimaging (NIC), and NP Cores. As part of Aim 5, we will share biosamples with NCRAD and other research collaborative efforts within NYU (e.g. NYU Metabolomics Lab) and outside collaborators from other ADRC (e.g. Microbiome Project). Last, in Aim 6, we will collaborate with other ADRCs to harmonize and optimize biomarker assays across centers. In summary, the BC will perform state-of-the-art biofluid biomarker analyses within the new ATN framework, and work with the NIC and CC cores to help understand disease heterogeneity and stage transitions to SCD, MCI, and AD. Further, it will help harmonize multiple biomarker assays and develop scientific discovery of novel ultrasensitive biomarkers for AD.
|
0.958 |
2020 — 2021 |
Wisniewski, Thomas M |
P01Activity Code Description: For the support of a broadly based, multidisciplinary, often long-term research program which has a specific major objective or a basic theme. A program project generally involves the organized efforts of relatively large groups, members of which are conducting research projects designed to elucidate the various aspects or components of this objective. Each research project is usually under the leadership of an established investigator. The grant can provide support for certain basic resources used by these groups in the program, including clinical components, the sharing of which facilitates the total research effort. A program project is directed toward a range of problems having a central research focus, in contrast to the usually narrower thrust of the traditional research project. Each project supported through this mechanism should contribute or be directly related to the common theme of the total research effort. These scientifically meritorious projects should demonstrate an essential element of unity and interdependence, i.e., a system of research activities and projects directed toward a well-defined research program goal. |
Novel Approaches to Understand the Pathogenesis and Treat Alzheimer's Disease @ New York University School of Medicine
OVERALL PROGRAM SUMMARY/ABSTRACT In this P01 proposal entitled: ?Novel Approaches to Understand the Pathogenesis and Treat Alzheimer?s Disease?, we seek to gain a better understanding of the heterogeneous pathogenesis of AD and how it is influenced by apolipoprotein (apo) E isotypes. The apoE4 allele is the major genetic risk factor for late-onset AD and has been strongly associated with increased amyloid plaques deposition in brain parenchyma and advanced vascular amyloid pathology; as well as, enhanced A? oligomerization. ApoE is also involved in synaptic plasticity, glucose metabolism, mitochondrial function, and vascular integrity. Currently, there is no consensus on how different apoE genotypes contribute to the pathogenesis of AD. The interrelated studies proposed in the three projects of this P01 will help elucidate this complex role of apoE in AD. Hence this P01 is addressing an issue of great significance. We propose an integrated, multidisciplinary research endeavor that brings together investigators with an extensive history of successful collaboration, who have expertise in diverse areas including proteomics, bioinformatics, neuropathology, AD mouse models, immunology, µMRI, µPET, medicinal chemistry and biomarker studies. Across all projects we will apply our innovative proteomic methods (with the assistance of the proteomics/neuropathology Core B) and use of common AD models and behavioral assessments (with the assistance of the transgenic/behavioral Core C), along with state-of-the-art biomarker technology using SIMOA and P01 investigator developed µMRI methodologies, to ensure synergism across all P01 studies. Scientific rigor of the P01 will be ensure by the Biostatistics and Bioinformatics Core (Core D). The three projects of this P01 are focused on the differential role apolipoprotein E (apoE) isoforms play in: 1) AD plaque and vessel amyloid development as assessed by unbiased proteomics across the full spectrum of AD pathology (Project 1); 2) innovative therapeutic approaches that target the A?/apoE interaction (Project 2); and 3) responses to our novel therapeutic immunomodulation that targets abnormal conformation (Project 3). Combined our efforts are anticipated to enhance our understanding of the differential effects of apoE isotypes on AD pathogenesis and accelerate the discovery of effective therapeutic approaches that address these diverse roles.
|
0.958 |
2020 — 2021 |
Wisniewski, Thomas M |
P30Activity Code Description: To support shared resources and facilities for categorical research by a number of investigators from different disciplines who provide a multidisciplinary approach to a joint research effort or from the same discipline who focus on a common research problem. The core grant is integrated with the center's component projects or program projects, though funded independently from them. This support, by providing more accessible resources, is expected to assure a greater productivity than from the separate projects and program projects. |
Alzheimer's Disease Research Center @ New York University School of Medicine
Since its inception in 1990, the Alzheimer's Disease Research Center (ADRC) at New York University Langone Health has facilitated pioneering research to define transitions from normal aging to the subjective cognitive decline (SCD), mild cognitive impairment (MCI), and early dementia stages of AD; as well as AD biomarker development. Here we propose to continue this long-standing successful research direction with a focus on understanding disease heterogeneity and delineating biomarkers and their role in these transitions, with the long-term goal of helping to develop novel interventions that will delay or prevent cognitive decline. The NYU ADRC has built an infrastructure that supports innovative research on AD and related dementias (ADRD) to help achieve the NAPA goal of a cure by 2025. This will be facilitated by our nine highly successful and interactive Cores (Admin; Clinical; Data Management & Statistical [DMS]; Neuropathology; Outreach, Education & Engagement [ORE]; Neuroimaging; Biomarker; Psychosocial; and Research Education Component [REC]). Together, our highly integrated cores will achieve the following aims: Aim 1. Enhance the performance of innovative research in ADRC by maintaining nine cores that focus on delineating biomarkers of the transitions from normal aging to SCD, MCI, and early dementia, and determining their roles to help develop novel interventions that delay or prevent these transitions. We will also facilitate training in this area. Aim 2. Contribute to the national network of ADRCs by providing clinical data, autopsy diagnoses, neuroimaging and biosamples to NACC and NCRAD, as well as to other research community collaborative efforts in ADRD. Aim 3. Recruit and retain a diverse subject population from clinical and community settings, via the ORE and Psychosocial Cores, with concomitant engagement of the local scientific and lay community in ADRD with seminars, poster sessions and the developmental projects via the Admin, ORE, and REC Cores. Aim 4. Foster the development of novel avenues of investigation with methodological developments by the cores (via innovative cognitive assessments, neuroimaging techniques, biomarkers and proteomic approaches), and encourage, recruit, and select developmental projects. Aim 5. Accelerate translational research across the ADRD spectrum by using biomarkers to better define the underlying disease heterogeneity and foster the development of novel therapeutic interventions that consider this heterogeneity. Aim 6. Facilitate the education and training of a diverse ADRD workforce. Our Center will enhance the scientific community's understanding of ADRD and expand the next generation of diverse ADRD scientists, via combined efforts of the Admin, ORE, and REC Cores. In summary, the NYU ADRD has facilitated pioneering research that defined the stage transitions from normal aging to dementia, and contributed to AD biomarker development from its inception. In the next five years of funding, this focus will be expanded by: 1) improving our understanding of disease heterogeneity; 2) identifying new biomarkers that will allow early detection; and 3) fostering research that will develop effective therapeutic interventions.
|
0.958 |