University of Arizona | Cellular & Molecular Medicine

Human Stem Cells & Computational Biology

We use patient-derived iPSC-cardiomyocytes, single-cell genomics, and computational approaches to understand cardiac disease mechanisms and identify therapeutic targets.

The Churko Lab

The Churko Lab at the University of Arizona integrates human iPSC technology with single-cell genomics, computational biology, and machine learning to understand the molecular basis of inherited cardiomyopathies. We develop new approaches for disease modeling, biomarker discovery, and therapeutic target identification.

Our foundation builds on methods pioneered at Stanford University in the laboratory of Dr. Joseph Wu, where we established approaches for iPSC-cardiomyocyte differentiation and high-throughput drug screening platforms that predict cardiotoxicity using patient-derived stem cells.

Biography

Dr. Jared Churko is an Associate Professor in the Department of Cellular and Molecular Medicine at the University of Arizona. He completed his postdoctoral training at Stanford University in the laboratory of Dr. Joseph Wu, where he developed methods for iPSC-cardiomyocyte differentiation and high-throughput cardiotoxicity screening. His research integrates human induced pluripotent stem cell technology with single-cell genomics and computational approaches to model inherited cardiac diseases, including arrhythmogenic cardiomyopathy, dilated cardiomyopathy, and atrial fibrillation. He serves on the editorial boards of JMCC Plus and Circulation: Heart Failure, and is a reviewer for NIH study sections.

Graduate Program Affiliations:

Biomedical Engineering Bio5 Institute Genetics GIDP Physiological Sciences GIDP

Research Focus

We combine stem cell biology, genomics, and computational methods to understand inherited cardiac diseases and identify new treatments.

Patient-Derived iPSC Models

We generate cardiomyocytes from patient blood to model inherited cardiac diseases including arrhythmogenic cardiomyopathy (PKP2, DSP), dilated cardiomyopathy (LMOD2, TTN), and atrial fibrillation. CRISPR/Cas9 isogenic controls enable precise genotype-phenotype analysis.

Single-Cell Transcriptomics

We use scRNA-seq to characterize cardiomyocyte heterogeneity during differentiation and identify disease-specific transcriptional signatures.

Multi-Omics Integration

Integrating RNA-seq, proteomics (mass spectrometry), and whole-genome sequencing to identify disease biomarkers and therapeutic targets.

3D Cardiac Tissues

Engineering multicellular constructs with cardiomyocytes, fibroblasts, endothelial cells, and epicardial cells for physiologically relevant models.

Integrated Platforms

From single-cell resolution to whole-genome analysis, our platforms generate deep biological insights.

Single-Cell RNA-Seq

Resolve cellular heterogeneity during cardiomyocyte differentiation. Identify rare cell populations and developmental trajectories at single-cell resolution.

10x Genomics Cell Lineage Trajectory Analysis

Bulk RNA-Seq & WGS

Comprehensive transcriptomic and genomic profiling to identify disease signatures, variant effects, and pathway dysregulation.

Differential Expression Variant Calling Pathway Analysis

Mass Spectrometry Proteomics

Quantitative proteomics to measure protein abundance, post-translational modifications, and protein-protein interactions.

TMT Labeling Phosphoproteomics Interactomics

3D Tissue Engineering

Multicellular constructs combining cardiomyocytes, endothelial cells, fibroblasts, and epicardial cells for physiologically relevant disease models.

Organoids EHT Co-culture

Developmental Cell Lineages

Using single-cell RNA-sequencing, we map the transcriptional landscape of cardiomyocyte differentiation to understand how cells acquire atrial versus ventricular identity. This work reveals the molecular programs driving cardiac specification and identifies markers for isolating specific subpopulations.

Disease Biomarker Discovery

We analyze patient-derived iPSC-cardiomyocytes using integrated multi-omics: whole-genome sequencing to identify variants, RNA-seq to measure transcriptional changes, and mass spectrometry to quantify protein alterations. This systems biology approach reveals disease mechanisms and therapeutic targets.

Bioengineered Disease Models

We engineer 3D cardiac tissues incorporating cardiomyocytes, fibroblasts, endothelial cells, and epicardial cells. These multicellular constructs better recapitulate the structural and functional properties of human myocardium, enabling more physiologically relevant disease modeling.

Expanding Beyond the Heart

In collaboration with the Center for Innovations in Brain Sciences, we apply our iPSC expertise to generate disease-specific neural cells for neurodegenerative disease research.

Alzheimer's Disease Parkinson's Disease ALS Multiple Sclerosis

Recent Publications

Published > 50 articles | 6,299 citations | h-index 32 | NIH iCite Weighted RCR 132.90 | mean RCR 2.83

2025

Modelling arrhythmogenic cardiomyopathy fatty-fibro pathology with PKP2-deficient epicardial cells derived from human iPSCs

Falana SL, Kazmouz SG, Iwanski JB, [...] Churko JM

Communications Biology

Senior Author iPSC Disease Modeling
2025

Minimal Component, Protein-Free, and Cost-effective Human Pluripotent Stem Cell Cardiomyocyte Differentiation

Iwanski JB, Lawal OS, Kwon WT, Vazquez I, Churko JM

Current Protocols

Senior Author Methods iPSC-CM
2025

Atrial Fibrillation Related Titin Truncation Is Associated With Atrial Myopathy in Patient-Derived iPSC Disease Models

Huang K, [...] Churko JM, [...] Laksman Z

Circulation: Genomic and Precision Medicine

iPSC Cardiomyopathy
2024

Leiomodin 2 neonatal dilated cardiomyopathy mutation results in altered actin gene signatures and cardiomyocyte dysfunction

Iwanski JB, Pappas CT, [...] Churko JM, Gregorio CC

NPJ Regenerative Medicine

Cardiomyopathy Disease Modeling
2023

Surfaceome mapping of primary human heart cells with CellSurfer uncovers cardiomyocyte surface protein LSMEM2

Luecke LB, [...] Churko JM, [...] Gundry RL

Nature Cardiovascular Research

Proteomics Heart Failure
2023

Epicardial placement of human placental membrane protects from heart injury in a swine model of myocardial infarction

Skaria RS, [...] Churko JM, [...] Konhilas JP

Physiological Reports

Cardiac Repair Translational
2017

High-throughput screening of tyrosine kinase inhibitor cardiotoxicity with human induced pluripotent stem cells

Sharma A, Burridge PW, McKeithan WL, [...] Churko JM, [...] Wu JC

Science Translational Medicine

Cardiotoxicity Drug Screening

Get in Touch

Interested in collaboration, joining the lab, or learning more about our research?

Location

Life Science North, Room 407
1501 N Campbell Ave
Tucson, AZ 85724