Awardees and Abstracts
2013 Awardees
Scholar Awards

In asthma, the muscle cells around the airways contract, narrowing the opening and reducing the flow of air in the lungs. As the disease progresses, the muscle cells enlarge and increase in number. These changes further increase the narrowing of airways and make asthma more difficult to treat. Dr. Deshpande is a pulmonary researcher trained in veterinary medicine and in molecular bioscience. Dr. Stephen Liggett from the University of Southern Florida and Dr. Deshpande made a curious and important discovery: muscle cells of the type that surround the airways (smooth muscle) express on their surface the same proteins that are used by the tongue to detect bitter foods. Remarkably, when these bitter food receptors on smooth muscle are activated, the muscle relaxes and airways dilate. Dr. Deshpande now wishes to assess the effect of these receptors on the size and proliferation of airway smooth muscle cells. His preliminary studies suggest that bitter taste receptors may block the proliferation of muscle cells. If so, this would enhance the potential of these receptors as a target for asthma therapy; activation of the receptors would both relax airway muscles and prevent their expansion.
Scientific AbstractTony Jun Huang, Ph.D.Pennsylvania State University (Now at Duke University)
Engineering Science and Mechanics
A Microfluidic System for Point-of-Care Immunophenotyping of Induced Sputum Samples from Asthmatic Patients
One of the goals of the AAF is to advance the techniques for assessing asthma and for identifying subsets of asthma. The importance of this approach was recently demonstrated by Dr. John Fahy and his colleagues at UCSF, who performed bronchoscopy on people with asthma and on people without asthma, obtaining samples of airway cells for study. By this approach, they were able to divide asthmatic patients into two large groups, those with airway inflammation and those without it. Only the patients with asthma responded to treatment targeting inflammation. Subsequent studies have suggested that asthma patients with airway inflammation can be further subdivided, based on the nature of the inflammation. The use of bronchoscopy to identify types of asthma, however, is not suitable for routine clinical practice, and the tests to identify different types of inflammation require complex equipment. Dr. Huang is an Associate Professor of Engineering at Penn State University, where he specializes in “microfluidics,” devices that allow the study of small samples of biological materials. Working with collaborators at the NIH, Dr. Huang has begun to develop small devices for analyzing small numbers of cells. In his AAF application, he proposes to adapt these techniques for the study of cells in sputum – material coughed up by patients with asthma. If this were successful it could be used both in research and in clinical care.
Scientific AbstractJing Liu, Ph.D.Northwestern University
Pulmonary and Critical Care Medicine
The Role of the Transcription Factor Miz1 in Asthma
The cells that line the airways are called epithelial cells. These cells were once thought to be just structural, i.e., they had no role in asthma. In the last decade, however, it has become clear that epithelial cells may play a central role in eliciting the inflammation of asthma. In fact epithelial cells may be the cells that first recognize certain allergens in the air, releasing chemicals that promote inflammation. Dr. Liu’s studies focus on the mechanisms by which this role for epithelial cells in asthma is regulated. In particular, she is studying a protein in these cells called Miz1, which dampens the ability of epithelial cells to elicit inflammation. Through understanding the molecular pathways by which Miz1 works, and how the activity of Miz1 itself is regulated, she seeks new targets to suppress inflammation in asthma. Toward this end, she will test her findings in a mouse model for asthma.
Scientific AbstractCharles D. Nichols, Ph.D.Louisiana State University Health Sciences Center, New Orleans
Pharmacology and Experimental Therapeutics
Serotonin 5-HT2A Receptor Activation As a Novel and Potent Therapeutic Strategy for Asthma
Dr. Nichols is a neurobiologist who studies molecules on the surface of nerve cells called serotonin receptors. These receptors alter the function of nerve cells when they are bound by a small molecule called serotonin. Serotonin receptors also bind other substances, however, one of them being the hallucinogenic drug LSD. Dr. Nichols was studying the effects of LSD and related drugs in rodents when he found that very low doses of these drugs – doses well below those that cause hallucinations – inhibit inflammation by activating the same specific serotonin receptors. When mice inhaled these drugs at low doses, they were resistant to asthma. For his AAF studies, Dr. Nichols will evaluate the activation of serotonin receptors as a therapy for treating asthma, using a mouse model for asthma.
Scientific AbstractPaula M. Oliver, Ph.D.University of Pennsylvania and Childrens' Hospital Philadelphia
Pathology
Activating E3 Ubiquitin Ligases to Treat Inflammation and Asthma
E3 ubiquitin ligases are enzymes that have broad effects in controlling the molecular pathways inside cells that lead to inflammation. Several E3 ubiquitin ligases suppress the inflammation that characterizes asthma. These ligases in turn depend on a group of small molecules in order to function. Dr. Oliver proposes to study these small molecules as potential targets for asthma therapy. The goal is to potentiate or to mimic their effects, activating ubiquitin ligases to suppress inflammation.
Scientific AbstractEric J. Sundberg, Ph.D.University of Maryland, Baltimore
Medicine & Institute of Human Virology
Manipulating Interleukin-33 Signaling to Control Asthma
There are different types of immune responses. The one most closely associated with asthma is called TH2 immunity, which promotes inflammation in asthma. TH2 immunity is activated in asthma when a protein called Interleukin-33 (IL-33) is released by the airway-lining cells. IL-33 binds to proteins on the surface of immune cells (IL-33 receptors), to promote TH2 immunity. Thus, IL-33 is itself a potential target for the treatment of asthma and so too are the IL-33 receptors. Dr. Sundberg is a structural biologist at the University of Maryland, where he has been studying a different aspect of the immune system activation (superantigens). He now proposes to turn his expertise to defining the structure of IL-33 when it is bound to its receptor. He will then use this structure to design molecules that will block the IL-33/receptor interaction, interrupting this pathway to TH2 immunity, providing a new approach to the treatment of asthma.
Scientific AbstractExtension Awards
Roderick MacKinnon, M.D.Rockefeller University & HHMI
Molecular Neurobiology and Biophysics
Drug Screen for Activators of the BK Channel for Asthma Treatment
In asthma, muscles around the airways tighten, narrowing the passage of air. The tightening of muscles is controlled by channels in the cell membrane that open to allow the release of potassium from the cell, relaxing the muscle. The most important current therapy in asthma, the inhalation of beta agonists (such as albuterol or salmeterol), ultimately works by opening potassium channels in muscle cells called BK channels. There are, however, no current therapies that act directly on BK channels. Thus, for his initial AAF Award, Dr. MacKinnon determined the structure of a large fragment of the BK channels so that drugs could be developed to open the channels. In the three years since the ending of his AAF Award, Dr MacKinnon has continued to work on this project, developing methods for producing the BK channel in quantities sufficient for the testing of drugs. In preliminary studies, he has screened 20,000 compounds and identified 9 that open the channel. He then compared features of these 9 compounds to identify structural themes. He now proposes to screen two different groups of compounds, each containing about 200,000 molecules. Potential drugs will undergo further testing for their capacity to relax smooth muscle cells, a new approach to opening the airways in asthma.
Scientific AbstractDaniele Piomelli, Ph.D.University of California, Irvine
Anatomy and Neurobiology
NAAA Inhibition as a Therapeutic Strategy for Asthma
Dr. Piomelli studies the role of fat molecules in inflammation. One group of these fats, called fatty acid ethanol amides (FAEs), acts inside inflammatory cells (macrophages) to inhibit their activity. As an outgrowth of his 2002 AAF Award, Dr. Piomelli discovered a way to increase FAEs in cells, thus decreasing inflammation. He achieved this by developing a new method for blocking the normal breakdown of FAEs in cells. He has continued to work on this project over the 8 years since his AAF Award ended. Importantly, he has developed a series of new drugs that are especially effective in blocking the breakdown of FAEs. Oral administration of these drugs to mice suppresses experimentally induced inflammation. Dr. Piomelli now proposes to test one of these drugs in mouse models of asthma. At the same time he will perform initial pharmacologic studies of the drug in rats (e.g., oral bioavailability, drug distribution, half-life). As Dr. Piomelli notes in his application, “the results will allow us to make a go-no go decision about the preclinical development of [this drug] in asthma.”
Scientific AbstractDeepak A. Deshpande, Ph.D., D.V.M. — 2013 Scholar Award
Thomas Jefferson University
Anti-mitogenic Effect of Bitter Taste Receptor Agonists on Airway Smooth Muscle
Airway remodeling characterized by increased airway smooth muscle (ASM) mass is a hallmark of obstructive diseases of airways such as asthma. Current asthma medications control inflammation and reverse airway obstruction effectively, yet have very limited effects in deterring airway remodeling. We identified the expression of bitter taste receptors (BTRs) on human ASM cells and activation with known agonists resulted in ASM relaxation. Aerosol challenge in normal and allergen sensitized- and challenged- mice resulted in a robust bronchodilation. These studies suggest that BTRs can be used as new therapeutic targets in asthma. To further establish their effectiveness as anti-asthma medication, we have proposed studies to determine their effects on ASM growth. Preliminary studies demonstrate that known BTR agonists inhibit mitogen-induced growth of healthy and asthmatic ASM cells. Based on these observations we hypothesize that BTR agonist inhibit ASM growth through novel anti-mitogenic signaling in ASM. Aim 1 studies will determine effects of BTR agonists on mechanisms that are involved in the growth (proliferation, size and survival) of normal and asthmatic human ASM. To characterize the signaling mechanism(s) studies in Aim 2 will determine effects of BTR agonists on mitogenic signaling pathways, and identify those effectors through which BTRs mediate their anti-mitogenic effects. Studies in Aim 3 will ascertain in vivo efficacy of BTR agonists in inhibiting ASM remodeling using house-dust mite challenged mouse model of allergic inflammation. Collectively, the findings from the proposed studies will identify a novel class of GPCRs agonists and signaling pathways that can mitigate ASM remodeling.
Tony Jun Huang, Ph.D. — 2013 Scholar Award
Pennsylvania State University (Now at Duke University)
A Microfluidic System for Point-of-Care Immunophenotyping of Induced Sputum Samples from Asthmatic Patients
Immunological phenotyping plays an important role in the development of approaches for personalized treatment of asthma. The objective of this project is to develop a point-of-care system that would allow automated, on-chip processing of induced sputum samples from asthmatic patients and analysis of immune cells present in sputum samples to establish a patient’s immunological phenotype.
The specific aims are:
Aim 1: Demonstrate a multi-color flow cytometry chip that can perform accurate, eight-parameter analysis of immune cells on samples of peripheral blood.
Aim 2: Develop a microfluidic chip for liquefying sputum samples .
Aim 3: Demonstrate an integrated microfluidic system for sputum processing and multi-parametric analysis of induced sputum samples from asthmatic patients, including the identification of neutrophils, lymphocytes, monocytes, eosinophils, epithelial cells, and macrophages.
The proposed microfluidic system have the following advantages: 1) eliminate the need for highly trained personnel; 2) assure consistency, accuracy, and reproducibility of the processing and analysis of induced sputum samples; 3) reduce time and cost for sputum processing and analysis; and more importantly 4) require a much smaller amount of sample for accurate analysis than current benchtop flow cytometry or other existing methods. The last feature is extremely important because the amount of cell samples available from some asthma patients is often not enough to run the conventional flow cytometry. This feature fills an unmet need in the field. With these advantages and unprecedented features, the proposed microfluidic flow cytometry system is expected to have a transformative impact on both asthma research and clinical care.
Jing Liu, Ph.D. — 2013 Scholar Award
Northwestern University
The Role of the Transcription Factor Miz1 in Asthma
Our long-tern goal is to understand the molecular mechanism underlying the pathogenesis of asthma and seek for novel therapeutic strategies. In this proposal, we propose to study the mechanism by which the transcription factor Miz1 controls the allergic response during asthma and its potential role in prevention and treatment of asthma.
Overwhelming evidence shows that Toll-like receptor 4 (TLR4)-mediated inflammatory response in airway epithelial cells (ECs) plays a central role in the development of allergy to a variety of environmental allergens. Recently, we discovered that Miz1 plays a critical role in modulating lipopolysaccharide (LPS)-induced, TLR4-mediated inflammation in lung epithelial cells. Loss of the transcriptional repression activity of Miz1 in lung ECs results in hyper-inflammation in intratracheal LPStreated mice, through transcriptional repression of C/EBPδ, an amplifier of LPS-induced inflammation. Most importantly, loss of the transcriptional repression activity of Miz1 promotes airway inflammation in a murine model of asthma induced by house dust mites (HDM). Based on this observation, we propose the studies to test whether Miz1 plays a critical role in regulation of the allergic response during asthma.
This proposal is novel, as it will study how Miz1 controls the allergic sensitization during asthma and how Miz1 itself is regulated during the pathogenesis of asthma. These studies will put forward a new paradigm regarding the molecular mechanism underlying the allergic sensitization during asthma, which has significant clinical implications.
Roderick MacKinnon, M.D. — 2013 Extension Award
Rockefeller University & HHMI
Drug Screen for Activators of the BK Channel for Asthma Treatment
High conductance voltage and calcium activated potassium channels (BK channels) control airway smooth muscle contractility by suppressing increases in intracellular calcium through negative feedback on voltage activated calcium channels ((1);(2);(3);(4)). It is known that the action of bronchodilators commonly used to treat asthmatic patients, such as β2 agonists and phosphodiesterase inhibitors, is suppressed when BK channels are inhibited (2). This suggests that BK channel activation is required to achieve the full therapeutic effect of these agents. In addition, human genetic studies link reduced BK channel activity to a familial form of asthma (5). Thus, every indication points to BK channel activators as a potential new therapy for asthma. Currently, the pharmaceutical industry relies on a cell-based assay using a thallium dye to identify BK channel active compounds. We have developed a new optical assay that is based on the expression and purification work originally supported by the AAF grant. Our new assay is not cell based but rather is analogous to an enzyme assay: therefore compounds identified must act directly on the channel and not a linked process within the cell. We have adapted the assay to 384 well plates for high-throughput analysis using a Hamamatsu 384 well plate reader, have identified our first novel activators of the BK channel, and have confirmed their activity in electrophysiological assays. Funding is requested to carry out a more extensive library screen and initial evaluation of compounds using electrophysiological and smooth muscle mechanics measurements, to begin structure-function chemistry on compounds already identified, and ultimately to attract the attention for further development by the pharmaceutical industry.
Charles D. Nichols, Ph.D. — 2013 Scholar Award
Louisiana State University Health Sciences Center, New Orleans
Serotonin 5-HT2A Receptor Activation As a Novel and Potent Therapeutic Strategy for Asthma
We have discovered that activation of the serotonin 5-HT2A receptor with the agonist (R)-DOI super-potently prevents the development of allergic asthma in a mouse model. This includes prevention of the development of airway hyperresponsiveness, eosinophilia, pulmonary inflammation, mucus overproduction, and inflammatory cytokine production from Th2 and innate cells within the lung. In this work we will investigate three aims towards bringing this novel therapeutic strategy to the clinic. In Aim 1, we will test the therapeutic efficacy of (R)-DOI to treat already established asthma, using a chronic OVA challenge mouse model. In Aim 2, we will test the efficacy of psilocybin to block the development of allergic asthma symptoms in an OVA challenge and sensitization rat model. If psilocybin is found to block asthma related symptoms similar to (R)-DOI, then moving forward to human clinical trials with psilocybin will be much more rapid because this drug is already in clinical trials for other indications. In Aim 3, we will perform a genetic association study between defined polymorphisms within the 5-HT2A receptor gene and multiple symptoms and variables of asthma. These results will inform personalized treatment strategies in the clinic to identify patients that may be more (or less) responsive to treatment with (R)-DOI or psilocybin. When all three aims are successful, we will have demonstrated the effectiveness of (R)-DOI and/or psilocybin to treat pre-existing asthma, be able to determine which patients may be more responsive to this treatment, and perhaps move quickly to clinical trials with psilocybin.
Paula M. Oliver, Ph.D. — 2013 Scholar Award
University of Pennsylvania and Childrens' Hospital Philadelphia
Activating E3 Ubiquitin Ligases to Treat Inflammation and Asthma
Asthma is one of the most common chronic illnesses affecting children in the U.S. and is increasing in prevalence worldwide. Genome wide association studies have implicated genes that regulate TH2 differentiation and function as well as genes that are involved in recognizing or responding to gut microbiota. E3 ubiquitin ligases regulate both of these processes. We have identified an E3 ubiquitin ligase adaptor, Nedd4-family interacting protein 1 (Ndfip1) that activates E3 ubiquitin ligases to dampen TH2 differentiation and limit proinflammatory cytokine production by macrophages. Importantly, we have identified a region of Ndfip1 that promotes ubiquitin charging of the E3 ubiquitin ligase Itch, thus triggering its activity.
We hypothesize that therapeutic mimics of Ndfip1 will activate E3 ubiquitin ligases such as Itch and thus limit proinflammatory cytokine production in T cells and macrophages. When these regulatory mechanisms are defective, such as in mice lacking Ndfip1, severe inflammation develops in the lung that resembles asthma, with goblet cell hyperplasia, accumulation of mucous in the airways and infiltration of TH2 cells and eosinophils. Additionally, we have identified polymorphisms in Ndfip1 that occur more commonly in patients with asthma.
Our Aims for this proposal are to (Aim 1) reveal the underlying biochemistry of how Ndfip1 activates Itch, and test whether Ndfip1 peptides can be used to dampen cytokine production by T cells (Aim 2) and macrophages(Aim 3) and thus limit inflammation. These studies will determine whether Ndfip1 peptide mimics have the potential to be used therapeutically for treating asthma.
Daniele Piomelli, Ph.D. — 2013 Extension Award
University of California, Irvine
NAAA Inhibition as a Therapeutic Strategy for Asthma
Identifying points of control in inflammation is essential to discover innovative anti-inflammatory medicines. The fatty acid ethanolamides (FAEs) are a family of endogenous anti-inflammatory lipid mediators. They are hydrolyzed by the enzyme N-acylethanolamine acid amidase (NAAA), which is highly expressed in macrophages. Funding from AAF allowed us to discover the first potent NAAA inhibitor, (S)-OOPP, and show that this compound increases FAE levels in macrophages and, by doing so, blunts inflammatory responses. These results suggest that NAAA might provide a novel target for anti-inflammatory medicines. Because (S)-OOPP has severely limited plasma stability, we modified its chemical scaffold and identified a novel class of potent, selective and drug-like NAAA inhibitors. In the present application, we propose to explore the utility of the prototype of this class, ARN726, as a therapeutic in asthma. Specifically, we will:
Aim 1: Characterize the anti-inflammatory properties of ARN726 in models of lung inflammation. Initial studies have shown that ARN726 is potent at inhibiting carrageenan-induced inflammation in mouse lungs. We will examine the consequences of oral administration of ARN726 in three additional mouse models of lung inflammation: (a) LPS-induced injury; (b) ovalbumin-induced inflammation; and (c) house dust mite-induced inflammation.
Aim 2: Evaluate the pharmacokinetic properties of ARN726. We will characterize the pharmacokinetic profile of oral and intravenous ARN726.
The proposed experiments will allow us to determine whether to continue our development work on ARN726 for asthma. If successful, they will provide preliminary data necessary to apply for an NIH STTR grant (Phase 2) to support such work.
Eric J. Sundberg, Ph.D. — 2013 Scholar Award
University of Maryland, Baltimore
Manipulating Interleukin-33 Signaling to Control Asthma
Asthma is an inflammatory airway disorder primarily associated with T helper type 2 (TH2) cell-mediated production of IgE antibodies and recruitment of mast cells and eosinophils. Interleukin (IL)-33 is a key activator of TH2 inflammatory responses in airways – when IL-33 binds its exclusive receptor ST2 on the surface of TH2 cells, it triggers a cascade of additional cytokines (e.g., IL-5, -9 and -13) that induce B cell isotype switching to IgE, as well as maturation and survival of mast cells and eosinophils. The genes for IL-33 and ST2 have been linked to asthma by genome-wide association studies and depletion of either of these proteins in model systems attenuates airway inflammation. Accordingly, targeted inhibition of IL-33-mediated TH2 responses represents an attractive, yet unrealized, therapeutic approach for the control of asthma. In the proposed studies, we will define the molecular mechanisms by which IL-33 and related cytokines agonize and antagonize cell signaling in order to rationalize our engineering efforts to develop a diverse set of IL-33 signaling inhibitors. Specifically, we aim to: (1) to elucidate the molecular basis of agonist and antagonist cytokine signaling through the IL-36 receptor by X-ray crystallography, mutagenesis and functional analyses (IL-33, an ST2 agonist, has no known antagonist counterpart); (2) to determine the structure of the IL-33 signaling complex and to engineer extracellular protein-based inhibitors of IL-33; and (3) to define the physical basis of cytoplasmic protein assembly initiated by IL-33 signaling complex formation and to develop intracellular peptide-based IL-33 inhibitors.