Initiatives

The Division of Nephrology in the Department of Medicine has a multitude of research initiatives currently underway.

Evren Azeloglu,PhD
Kidney Precision Medicine: Systems Bioengineering Lab studies cellular mechanobiology using combination of multi-omics technologies, quantitative microscopy, and stem cell-based tissue engineering methods.

Babu Padanilam, PhD
We are studying the mechanisms of cellular injury in acute kidney injury (AKI) and renal fibrogenesis in chronic kidney disease (CKD) models. The molecular mechanisms leading to energy depletion and cell death in the kidney proximal tubules, are investigated in various experimental models of AKI. Significant risks for long term loss of renal function and development of renal fibrosis and progression to CKD exists after AKI. Current projects include delving into the regulation of fatty acid oxidation in AKI, role of sympathetic nerve activity, neural factors and epoxyeicosatrienoic acids (EETs) in the regulation of renal fibrogenesis in different models of CKD and developing novel drug delivery approaches in AKI and CKD.

Kirk Campbell, MD
Our research program seeks to define key signaling pathways essential for cellular homeostasis with the overarching goal of developing novel therapeutic options

Lili Chan, MD
Our research looks to use artificial intelligence to identify novel predictors of adverse clinical outcomes in patients with kidney disease.

Nicholas Chun, MD
Our lab uses transplant models to understand how the interaction between exosomes/extracellular vesicles and the complement system modulates immune responses with the hope of identifying novel therapeutic targets to improve outcomes for patients with immune-mediated diseases.

Steven G. Coca, DO, MS
Using biomarkers to improve risk-stratification or assess therapeutic response in acute and chronic kidney disease.

Paolo Cravedi, MD
The Cravedi Lab works on basic and translational research projects testing a) noninvasive biomarkers to guide immunosuppressant treatments in kidney transplant recipients, b) associations between T cell exhaustion markers and allograft function, c) protolerogenic mechanisms of erythropoietin (EPO) in transplantation and kidney diseases, and d) the role of complement system in non-antibody mediated kidney diseases. 

Ilse Daehn, MD
Exploring the complexity of signaling crosstalk between cells in the kidney. Examining molecular mechanisms in the glomerulus that result in kidney disease progression and diabetic nephropathy to identify novel glomerular lesion-specific therapeutic targets and biomarkers.

Miguel Fribourg, PhD
We are studying the mechanisms of cellular injury in acute kidney injury (AKI) and renal fibrogenesis in chronic kidney disease (CKD) models. The molecular mechanisms leading to energy depletion and cell death in the kidney proximal tubules, are investigated in various experimental models of AKI. Significant risks for long term loss of renal function and development of renal fibrosis and progression to CKD exists after AKI. Current projects include delving into the regulation of fatty acid oxidation in AKI, role of sympathetic nerve activity, neural factors and epoxyeicosatrienoic acids (EETs) in the regulation of renal fibrogenesis in different models of CKD and developing novel drug delivery approaches in AKI and CKD.

Luca Gusella, PhD
Pathogenetic mechanisms underlying Autosomal Dominant Polycystic Kidney Disease. Focusing on the early events of cystogenesis and the molecular and cellular responses immediately dependent on the dysregulation of PKD1 or PKD2.

John Cijiang He, MD, PhD
The research emphasis in the joint He and Lee laboratories are focused on uncovering the molecular mechanisms that govern the initiation and progression of chronic kidney disease of various etiologies for optimal therapeutic targeting. The disease areas include DKD, immune-mediated GN, FSGS, HIV-mediated CKD, and kidney fibrosis.

Pathogenesis of HIV nephropathy
Using systems biology approach to identify signaling pathways involved in HIV kidney disease and identifying new drugs and drug targets for treatment of kidney disease

Podocyte biology and pathology
Role of retinoic acid as a therapy for glomerular disease with podocyte injury

Kidney fibrosis:
Validation of HIPK2 as a potential anti-fibrosis drug target 

Lewis Kaufman, MD
The Kaufman lab investigates molecular pathways that govern glomerular injury in chronic kidney disease development. Our goal is to devise novel cell-targeted therapeutic approaches that protect glomerular cells and mitigate chronic kidney disease progression.

Kristin Meliambro, MD
Our lab utilizes cell-based and murine models and clinical samples to define mediators of podocyte injury and glomerular disease progression.

Dinushika Mohottige, MD
Dr. Mohottige's research team is focused on identifying, measuring, and mitigating disparities in kidney care and kidney health outcomes. Using qualitative and quantitative methods, studies span from those examining structural/environmental inequalities associated with CKD risk, to those examining trust and other factors associated in non-standard kidney transplant acceptance. Fundamentally, these studies use patient and community centered approaches to enhance equity in kidney health.

Girish N Nadkarni, MD, MPH, CPH
Utilize electronic medical record data and banked biospecimens for risk stratification in patients with chronic kidney disease.

Joshua Rein, DO
Dr. Rein’s research, funded by a Veterans Affairs Career Development Award, explores how cannabis and cannabinoids affect kidney function given their widespread growing popularity despite an uncertain impact on health. His basic science research utilizing preclinical animal models examines the physiological impact of cannabinoids on total body fluid and electrolyte homeostasis and aims to identify future targets for novel therapeutics to treat disorders such as hyponatremia and volume overload. His clinical research focuses on the risks, benefits, and clinical implications of cannabis consumption among people with kidney disease and those at risk for developing kidney disease.

Rajeev Rohatgi, MD
The focus of the lab to study the interaction of renal cholesterol efflux pathways on the development of sodium sensitive hypertension and tubulo-interstitial inflammation. The lab utilizes a combination of murine models, molecular biology, single tubule isolation and electrophysiology to address this question.

Lisa M. Satlin, MD
The Satlin lab focuses on defining the mechanisms leading to the acquisition, maintenance and regulation of transepithelial transport in the mammalian distal nephront, responsible in the adult for the final renal regulation of total body K+ and Na+ homeostasis. Recent efforts have been devoted to developing model systems, including 3D bioprinted kidney tubules and human PSC-derived kidney organoids, which recapitulate ion transport and signaling phenotypes of the in vivo distal nephron. Her lab particpates as a national "single nephron physiology" Core of an NIH-funded Center for Kidney Research, providing external investigators access to microphysiologic techniques to define function of single tubules/cells in native tissue.

Jaime Uribarri, MD
Dr. Uribarri's research focuses in two main areas: 1) role of dietary advanced glycation endproducts (AGEs) and dietary phosphorus as risk factors for cardiovascular disease in CKD patients and 2) Better understanding of barriers to implement Home dialysis programs in ESRD patients.

Mon-Wei Yu, MD
Our research focuses on the role of complement proteins in acute kidney injury (AKI) and kidney fibrosis development using various models including primary cells, mouse, and kidney organoids.

Dr. Weijia Zhang
Our research is focused on: 1) Perform translational genomic analysis in the biopsy tissue and/or whole blood for biomarker and drug target discovery and molecular mechanistic studies of human complex diseases. 2) Develop AI-based tools for reliable and efficient assessment of pathological slides for diagnosis/prognosis or organ utilization guidance in kidney transplant or other diseases.