NIH Programs
The U.S. National Institutes of Health (NIH) has engaged Vala Sciences to complete the following research programs utilizing Vala’s technology platform. Additional detail for each project can viewed by clicking the NIH link button in each project’s description.
NIH Grants
Neuroscience
Project Number: AG062012
Project Title: The Alzheimer’s Therapeutics Screening Assay: a high-throughput drug-discovery platform utilizing neurons and microglia derived from human induced pluripotent stem cells and Kinetic Image Cytometry
Project Description:
No new drug for Alzheimer’s Disease has been approved since 2003, highlighting the need for more predictive preclinical drug discovery systems. This project aims to develop the Alzheimer’s Therapeutics Screening Assay (ATSA) using co-cultures of neurons, microglia, and astrocytes derived from human induced pluripotent stem cells. We will expose these cultures to Alzheimer’s-relevant stressors like amyloid beta oligomers, apolipoprotein E-isoform 4 fragments, and excitotoxic agents. We will then assay each cell type for changes in morphology, synapses, and calcium and voltage activity. The results from this research will enable the ATSA to identify compounds that ameliorate Alzheimer’s neurotoxicity.
Funding agency: NIH/National Institute on Aging
Award Date: September 2018
Project Number: MH119621
Project Title: The Microscopic Imaging of Epigenetic Landscape-NeuroDevelopment (MIEL-ND) assay: a high throughput platform to screen compounds for neurodevelopmental effects
Project Description:
Recent research suggests that neurodevelopmental diseases (Autism Spectrum Disorder, intellectual disabilities, bipolar disorders, schizophrenia, etc.) can result from disruption of epigenetic histone and DNA modification by prenatal exposure to environmental toxicants. This project will develop the Microscopic Imaging of Epigenetic Landscape – NeuroDevelopment (MIEL-ND) assay, which will enable testing of chemicals for epigenetic effects on neurodevelopment. The MIEL-ND assay will use immunolabeling, automated high-throughput imaging, and cell-by-cell analysis to screen chemicals in the US EPA ToxCast program for effects epigenetic modification patterns in human neural precursor cells. The assay will also screen for effects on neurite and synapse formation in neurons derived from human induced pluripotent stem cells. The MIEL-ND assay will provide a less expensive, higher throughput, and more predictive alternative to animal testing.
Funding agency: NIH/National Institute of Mental Health
Award Date: September 2018
Project Number: ES026268
Project Title: Assay of environmental toxicants for toxicity related to Alzheimer’s Disease utilizing human iPSC-derived- neurons and microglia
Project Description:
Particulate air pollution 2.5 microns in diameter or less (PM2.5 particles) can enter the human body through the respiratory system and directly or indirectly damage the brain. High concentrations of PM2.5 particles in heavily polluted areas have been linked to increased Alzheimer’s Disease incidence. This project aims to increase our understanding of how PM2.5 particles cause neurodegeneration. We will investigate the effects of PM2.5 particles on neurons and microglia derived from human induced pluripotent stem cells. We will assay cells with multiple genotypes, including those expressing the epsilon-4 variant of apolipoprotein E, which increases risk of late-onset Alzheimer’s and likely increase susceptibility to environmental toxicants. This assay system will also be useful in identifying therapeutics for Alzheimer’s and other neurodegenerative diseases.
Funding agency: NIH/National Institute of Environmental Health Sciences
Award Date: September 2018
Project Number: ES026268
Project Title: Assay of chemicals for Parkinson’s toxicity in human iPSC-derived neurons
Project Description:
Environmental toxicants like rotenone, paraquat, or maneb can increase risk of Parkinson’s Disease. This project aims to develop an assay system to screen environmental toxicants for potential Parkinson’s toxicity. Our assay will detect toxicity in dopaminergic neurons (the main cell type affected by Parkinson’s) and microglia derived from human induced pluripotent stem cells. We will test each toxicant for effects on Parkinson’s-associated protein aggregation, mitochondrial function, and calcium and voltage activity. The resulting assay system will enable comprehensive and clinically relevant screening for toxicants with potential Parkinson’s-inducing effects.
Funding agency: NIH/National Institute of Environmental Health Sciences
Award Date: September 2015
Project Number: AG062012
Project Title: The Alzheimer’s Therapeutics Screening Assay: a high-throughput drug-discovery platform utilizing neurons and microglia derived from human induced pluripotent stem cells and Kinetic Image Cytometry
Project Description:
No new drug for Alzheimer’s Disease has been approved since 2003, highlighting the need for more predictive preclinical drug discovery systems. This project aims to develop the Alzheimer’s Therapeutics Screening Assay (ATSA) using co-cultures of neurons, microglia, and astrocytes derived from human induced pluripotent stem cells. We will expose these cultures to Alzheimer’s-relevant stressors like amyloid beta oligomers, apolipoprotein E-isoform 4 fragments, and excitotoxic agents. We will then assay each cell type for changes in morphology, synapses, and calcium and voltage activity. The results from this research will enable the ATSA to identify compounds that ameliorate Alzheimer’s neurotoxicity.
Funding agency: NIH/National Institute on Aging
Award Date: September 2018
Cardiac Biology
Project Number: ES023521
Project Title: Stem cell toxicology assays for cardiac differentiation
Project Description:
Environmental toxicants can disrupt fetal heart formation and lead to miscarriages or congenital heart defects. Little is known about how such chemicals might affect early heart development. This project aims to develop a Cardiopoiesis (heart formation) Assay to screen for compounds that influence the differentiation of cardiomyocyte precursors from multipotent mesodermal progenitors. The Cardiopoiesis Assay will test for compound effects on the ability of multipotent mesodermal progenitors derived from human induced pluripotent stem cells to express MYH6, a cardiac-specific gene, or markers of alternative cell fates. This assay will provide a less expensive, higher throughput, and more predictive alternative to animal testing.
Funding agency: NIH/National Institute of Environmental Health Sciences
Award Date: September 2013
Project Number: HL112521
Project Title: Optogenetic Multiparametric Assay for HT Cardiotoxicity Testing
Project Description:
Many drug candidates fail in clinical trials or after market launch due to cardiotoxicity, highlighting the need for more predictive preclinical cardiosafety screens. This project aims to develop a high throughput cardiosafety screening platform using cardiomyocytes derived from human induced pluripotent stem cells, which exhibit the contractile phenotype of human ventricular cardiomyocytes. Following compound exposure, we will use automated microscopy and image analysis to quantify calcium and voltage transients that occur with each cardiomyocyte contraction. We will then fix and immunolabel the cells for atrial and ventricular markers and correlate cell identity to activity on a cell-by-cells basis. This project will enable high throughput, accurate, and cost-effective cardiosafety testing.
Funding agency: NIH/National Heart, Lung, and Blood Institute
Award Date: March 2012
Project Number: HL086076
Project Title: Live cell and HCS assays to quantify production of cardiomyocytes from stem cells
Project Description:
Cardiomyocytes derived from embryonic stem cells may restore cardiac function in patients suffering from heart failure. This project aims to develop a high content screening system to measure the efficiency of cardiomyocyte production from embryonic stem cells. The system will record intracellular calcium transients, which are a hallmark of differentiated cardiomyocytes. The system will also quantify expression and localization of cardiac-specific structures like myofibrils and proteins like SERCA2. The system will enable testing of candidate compounds for their ability to influence the differentiation of cardiomyocytes for treatment of heart failure.
Funding agency: NIH/National Heart, Lung, and Blood Institute
Award Date: September 2006
Mitochondrial Function
Project Number: HL105061
Project Title: A New Toxicity Screen to Assess Mitochondrial DNA Content and Protein Synthesis
Project Description:
Many hepato- and cardiotoxic drugs and drug candidates act as mitochondrial toxins. While several assays exist to measure acute mitochondrial toxicity (changes in membrane potential, mitochondrial membrane integrity, and reactive oxygen species generation), few assays can detect long term effects such as mitochondrial DNA depletion and inhibition of mitochondrial protein synthesis. This project aims to develop an automated high content image analysis assay that can identify compounds that deplete mitochondrial DNA and/or proteins (e.g., electron transport chain subunits). Our assay will use automated image analysis to quantify mitochondrial DNA and proteins in fluorescent cell images following compound exposure. We will validate the assay in hepatocyte and cardiac model systems using compounds known to deplete mitochondrial DNA and proteins. Our assay will provide a high throughput, cost-effective solution to screen for long term mitochondrial toxicity in the early stages of drug development.
Funding agency: NIH/National Heart, Lung, and Blood Institute
Award Date: August 2010
Skeletal Muscle
Project Number: AR073565
Project Title: The Stem Cell-Derived Muscle Function Assay: A High Throughput Screening Platform Utilizing Kinetic Image Cytometry to Discover Therapeutics for Duchenne Muscular Dystrophy
Project Description:
Duchenne Muscular Dystrophy (DMD) is an early onset, progressive, and fatal disease caused by mutations in the DMD gene that prevent expression of the skeletal muscle structural protein dystrophin. This project aims to develop the Stem Cell-derived Muscle Function Assay (SCMFA), an in vitro high throughput screening system for potential DMD therapeutics. The SCMFA will use skeletal muscle differentiated from human pluripotent stem cells from healthy subjects, subjects with DMD, or subjects with Becker Muscular Dystrophy, a milder skeletal muscle condition. We will develop methods to assay the effects of paced contraction and stretching on skeletal muscle function and contractile apparatus biomarker expression. The methods developed for the SCDMFA will apply to other inherited muscular dystrophies and related conditions.
Funding agency: NIH/National Institute of Arthritis and Musculoskeletal and Skin Diseases
Award Date: September 2018
Project Number: AR055604
Project Title: Automated Analysis of Skeletal Muscle Fiber Cross-sectional Area and Metabolic Type
Project Description:
Skeletal muscle morphology provides insight into a wide variety of health issues such as aging, muscle denervation, muscular dystrophy, nutrition, and exercise physiology. This project aims to develop automated image analysis software that can measure the cross-sectional area of muscle fibers in skeletal muscle tissue sections. We will also immunolabel the tissue sections for myosin type I to quantify the percentage of slow twitch fibers within each muscle. The proposed research will generate a high throughput assay that can quantify the effects of experimental interventions in altering muscle physiology and health.
Funding agency: NIH/National Institute of Arthritis and Musculoskeletal and Skin Diseases
Award Date: July 2008
Liver
Project Number: DK082087
Project Title: Automated quantification of lipid droplets in fatty liver tissue sections
Project Description:
Non-alcoholic fatty liver disease results from lipid accumulation in liver cells (hepatocytes) followed by cell damage or death, inflammation, and cirrhosis. This project aims to develop an assay to detect changes in hepatocyte lipid content. We will develop image analysis techniques to quantify the frequency and size of hepatocyte lipid droplets in images of human liver tissue sections. Our assay will enable high content screening for potential therapies for non-alcoholic fatty liver disease and other metabolic conditions.
Funding agency: NIH/National Institute of Diabetes and Digestive and Kidney Diseases
Principal Investigator: Patrick M. McDonough
First Budget Start Date: March 2010
Project Number: DA026213
Project Title: A High Throughput Imaging Assay for Hepatic Lipid Droplet Formation
Project Description:
Non-alcoholic fatty liver disease results from lipid accumulation in liver cells (hepatocytes) followed by cell damage or death, inflammation, and cirrhosis. This project aims to develop a high content screening assay to identify compounds that alter hepatic lipid droplet formation in hepatocyte-derived cell lines (murine AML-12 cells and human HuH-7 cells). Our assay will enable discovery of new agents that can treat non-alcoholic fatty liver disease and other metabolic conditions.
Funding agency: NIH/National Institute on Drug Abuse
Award Date: June 2008
Project Number: DK074333
Project Title: HT Image Assay of Lipid Droplet Formation in Human Adipocytes
Project Description:
Dysregulation of adipocyte differentiation and lipid metabolism can contribute to obesity. This project aims to develop an image-based high content screen to test the effects of drug candidates on the differentiation and metabolic state of cultured human adipocytes. The screen will use image analysis software to quantify the number, size, and distribution of lipid droplets in maturing human adipocytes. The screen will also quantify the expression and distribution of proteins involved in lipid droplet formation and metabolism such as perilipin, adipophilin, and hormone sensitive lipase. Our screen will identify compounds with potential to treat obesity and related conditions like non-alcoholic fatty liver disease, diabetes, and heart disease.
Funding agency: NIH/National Institute of Diabetes and Digestive and Kidney Diseases
Award Date: July 2006
Cancer
Project Number: MH082378
Project Title: High Throughput Imaging Assay for Beta-Catenin
Project Description:
In normal cells, β-catenin binds transmembrane proteins like cadherins at the plasma membrane. In tumor cells, however, β-catenin moves to the nucleus, where it interacts with transcription factors to increase the expression of proteins that promote mitosis and tumor growth. This project aims to develop an assay to identify compounds that change β-catenin expression and cellular distribution in HeLa cells. The assay will use automated image acquisition and analysis to quantify the amount of β-catenin at the membranes and nuclei of treated cells. The assay will identify potential cancer therapeutics as well as treatments for other conditions that result from β-catenin dysregulation, such as depression and dementia.
Funding agency: NIH/National Institute of Mental Health
First Budget Start Date: August 2007
Pancreas
Project Number: DK076510
Project Title: Development automated assay-regulators insulin synthesis
Project Description:
Insulin promoter activity is a key indicator of pancreatic beta-cell function. Compounds that stimulate or maintain insulin expression have the potential to preserve and enhance beta-cell function in diabetes. This project aims to develop an automated screen for small molecules that modulate insulin expression in mouse pancreatic MIN6 cells. The system will use automated digital microscopy and image analysis algorithms to measure the expression of a fluorescent reporter under the control of the insulin promoter. This screen can identify potential diabetes therapeutics as well as compounds that inhibit insulin promoter activity and may cause beta-cell dysfunction.
Funding agency: NIH/National Institute of Diabetes and Digestive and Kidney Diseases
Award Date: September 2006
NIH Contracts
Contract Number:75N91019C00030
Contract Title:The Microscopic Imaging of Epigenetic Landscape Breast Tumor Differentiation (MIEL-BTD) Assay: A High Throughput Screen to Identify Novel Therapeutics for Breast Cancer
Contract Research Summary:
Compounds that induce differentiation of tumor cells towards a quiescent, drug-sensitive phenotype have the potential to improve breast cancer treatment. This contract research developed an imaging-based high content screen for effects on epigenetic histone modifications, which control gene expression and differentiation state. We demonstrated that Vala’s Structured Illumination MicroscopyTM significantly improves the imaging and analysis of histone modification patterns versus standard widefield microscopy. We performed a pilot 12-compound screen that identified several compounds that affect histone modification patterns. To measure the effects of epigenetic changes, developed a CyteSeer® image analysis algorithm to quantify compound effects on lipid droplet formation, a key biomarker of breast cell differentiation. We also produced MCF-7 breast cancer cell spheroids in 384-well plates to simulate compound effects in the tumor microenvironment. Future research will use genetically encoded epigenetic probes to image and analyze the distribution of epigenetic histone tags in breast cancer tumor spheroids.
Funding Agency: NIH/National Cancer Institute
Contract Period: September 2019 – June 2020
Contract Number: HHSN261201400025C
Contract Type: Phase I
Contract Title: In vitro assay of the initiation of breast cancer metastasis
Contract Research Summary:
Metastasis, the migration of tumor cells to distant tissues to form new tumors, drives breast cancer progression. However, few therapies exist that can inhibit this process. In this contract research, we developed an assay for breast cancer cell migration that can identify compounds with the potential to inhibit metastasis. We developed methods to model the breast tumor microenvironment by culturing MCF-7 and MDA-MB-231 cells in 3D tumor spheroids under hypoxic conditions. We also developed an assay using microchannel array plates to quantify tumor cell migration toward chemoattractants in normoxia or hypoxia and in the absence or presence of macrophages. In the future, we plan to conduct a high content screen using our assay to test potential chemotherapeutics for the ability to inhibit tumor cell migration and/or proliferation in our system.
Funding Agency: NIH/National Cancer Institute
Contract Period: September 2014 – June 2015
Contract Number: HHSN271201400029C
Contract Type: Fast-Track
Contract Title: A high throughput assay for neural crest cell migration and toxicity
Contract Research Summary:
Cell migration plays a key role in embryonic development and in cancer metastasis. In this contract research, we developed a high content screening system to test compound effects on cell migration. We designed a 384-well plate scratcher to clear small areas in cell monolayers. We also developed CyteSeer® image analysis algorithms to quantify cell migration into the cleared areas. Our assay consistently found that the actin polymerization inhibitor cytochalasin D inhibits cell migration in neural crest precursor cells, vascular endothelial cells, and MCF-7 breast cancer cells. We developed methods to multiplex our scratch assay with measurements of mitochondrial membrane potential and β-catenin nuclear localization. We developed a continuous scanning high content imaging system to enable imaging and analysis of more than 100,000 wells per day. This high content screening system can measure cell migration in a variety of biological systems.
Funding Agency: NIH/National Center for Advancing Translational Sciences
Contract Period: September 2013 – July 2016
Contract Number: HHSN261201100111C
Contract Type: Phase I
Contract Title: Automated karyometry as a companion diagnostic for chemoprevention of breast cancer
Contract Research Summary:
Karyometry is an image analysis method that extracts dozens of features describing nucleus size, shape, intensity, and chromatin distribution from tissue biopsies. Karyometry detects cancer-related nuclear abnormalities in breast tissue biopsies with high sensitivity, but its low throughput impedes breast cancer diagnosis and drug discovery. In this contract research, Vala Sciences and the Arizona Cancer Center used karyometry to confirm the beneficial effects of Arzoxifene, a selective estrogen receptor modulator, in women at high risk for breast cancer. We also used karyometry to predict which patients would respond to Arzoxifene with 70% accuracy. To develop a high-throughput karyometry-based assay, we established reference cell lines for normal and abnormal phenotypes and wrote CyteSeer® image analysis algorithms for automated recognition of nuclei and karyometric analysis. This project lays the foundation for future development of karyometry as a diagnostic test for the potential of drugs to prevent or reverse development of cancer.
Funding Agency: NIH/National Cancer Institute
Contract Period: September 2011 – June 2012
Contract Number:HHSN261201000088C
Contract Type:Fast-Track
Contract Title: A hapten based multiplexed assay for HER2/neu, ER, and PR in breast cancer tumors
Contract Research Summary:
Breast cancer cell expression of the HER2/neu growth factor receptor, the estrogen receptor, and the progesterone receptor can indicate malignancy and predict response to therapy. In this contract research, we developed a high content screen for these biomarkers using haptens (molecular tags that conjugate primary antibodies to secondary antibodies or other reagents) to enable simultaneous cell-by-cell quantification of all three biomarkers, cytokeratin staining, and nuclear ploidy. We also developed methods for hematoxylin/eosin staining for tissue morphology from the same tissue sections. We improved CyteSeer® automated nuclear recognition algorithms to segment crowded nuclei in breast cancer tissue sections. We validated our assay using a tissue micro array of eight reference breast cancer cell lines and samples from 75 patients, and our results had high concordance with conventional pathology phenotyping. Our assay can improve the throughput and accuracy of clinical breast cancer diagnosis.
Funding Agency: NIH/National Cancer Institute
Contract Period: September 2010 – September 2013