Division of Clinical Pharmacology

Research Centers and Labs

Research Centers

  • Autonomic Dysfunction Center: The Autonomic Dysfunction Center conducts clinical research studies to better understand the pathophysiology of the autonomic nervous system and its role in autonomic dysfunction, all the while seeking to discover new therapies to help optimize treatment of known autonomic disorders.
  • Vanderbilt Center for Arrhythmia Research and Therapeutics: An interdisciplinary center, directed by Bjorn Knollmann, MD, PhD, that focuses on understanding mechanisms predisposing to abnormal heart rhythms, and uses this information for optimal personalized patient care.
  • Vanderbilt Center for Bone Biology: A cross-disciplinary center within the Division of Clinical Pharmacology that studies basic and translational aspects of bone development, bone loss, fracture repair and metastatic disease to bone. Faculty from the Division of Clinical Pharmacology and departments of Pediatrics, Orthopedic Surgery and Biomedical Engineering participate in this center.
  • Vanderbilt Hypertension Center of Excellence: One of 13 American Heart Association’s centers of excellence focused on the diagnosis and treatment of humans with hypertension. Investigators in this center have discovered basic mechanisms underlying this common human disease, and have define novel causes of immune activation in hypertension and related cardiovascular illnesses.
  • Vanderbilt Institute of Chemical Biology

Research Labs

  • M. Alexander Lab – Focuses on the role of counter-regulatory immune mechanisms in the pathogenesis of hypertension and related end-organ damage, with the goal of co-opting these mechanisms for new therapies.
  • I. Biaggioni Lab – Studies mechanisms underlying dysregulation of autonomic function in humans. Dr. Biaggioni is the Director of the Vanderbilt Autonomic Dysfunction Center and oversees a large research program to improve the care of humans with autonomic dysfunction.
  • S. Dikalov Lab – Studies how reactive oxygen species are generated in mammalian cells and their role in pathophysiological conditions like hypertension. Dr. Dikalov's group has defined novel perturbations of mitochondrial function in diseases like hypertension, atherosclerosis and diabetes.
  • A. Dikalova Lab – Studies the role of mitochondrial proteins including the Sirtuins in modulating mitochondrial function. Dr Dikalova uses unique mouse models to delete sirtuins in target tissues to study how these affect target organ function in hypertension.
  • A. Gamboa Lab – Focuses on the study of the interactions between the autonomic nervous system and the mechanisms involved in the regulation of blood pressure, blood flow, and insulin sensitivity in obesity-associated hypertension.
  • J. Gamboa Lab – Studies factors that modulate mitochondrial function in humans. Dr. Gamboa's studies have elucidated perturbations of energy production and utilization in conditions like chronic renal disease, hemodialysis and systemic lupus.
  • Glazer Lab – Studies mutations that cause arrhythmia disorders and other diseases associated with ion channels. We use high-throughput in vitro methods and large cohort datasets to discover new disease-associated mutations.
  • D. Harrison Lab – Studies how the immune system and chronic inflammation are engaged in cardiovascular diseases. In particular, his group has defined novel links between oxidative injury and immune activation in hypertension and related conditions.
  • R. Johnson Lab – Focuses on mechanisms of breast cancer dissemination to bone, tumor recurrence in distant metastatic sites, and protecting skeletal health in the setting of tumor-targeted agents to prevent bone metastases and fractures.
  • Kirabo Lab– The overarching goal of the Kirabo Lab is to define mechanisms that lead to health disparities in cardiovascular disease. Dr. Kirabo's lab is working to define mechanisms that contribute to inflammation and autoimmunity in cardiovascular disease, with a particular focus on immune mechanisms of excess dietary salt-induced cardiovascular and kidney disease, and how this is modulated by the gut microbiota and viral infections including HIV/AIDS.
  • B. Knollmann Lab – Investigates molecular arrhythmia mechanisms involving alterations in the functioning of ion channels, myofilaments and calcium release channels. His lab is developing and testing new anti-arrhythmic therapies in human induced pluripotent stem cell models, in animal models and in humans.
  • B. Kroncke Lab – Aims to accurately predict the meaning of genetic variants to the individuals who carry them. Dr. Kroncke's approach is to collect as much phenotype information, at high and low resolution, as is feasible for the largest number of variants and then leverage high-throughput experimental and computational methods to fill in the details for yet uncharacterized variants.
  • J. Luther Lab – Focuses on the role of the renin-angiotensin-aldosterone system on glucose regulation using human and animal models. Dr. Luther’s group uses pharmacologic tools and genetic interruption to investigate the effects on insulin secretion and insulin sensitivity in vivo. Dr. Luther is also the director of the Vanderbilt AHA Comprehensive Hypertension Center, where he evaluates and treats resistant and secondary causes of hypertension such as primary aldosteronism and renovascular hypertension.
  • G. Milne Lab – Studies the metabolism of oxidized lipid mediators in health and human disease. The lab has a particular interest in the use of oxidized lipid mediator metabolites as biomarkers of their endogenous production. Dr. Milne is the Director of the Eicosanoid Core and the Neurochemistry Core, which each provide analysis and quantitation of these small molecules using mass spectrometry.
  • J. Mosley Lab – Focuses on identifying translational applications of polygenic variation to clinical medicine to improve health care delivery, reduce unnecessary health care utilization and improve risk stratification. This work typically leverages data from electronic health record (EHR) data sources and epidemiological studies.
  • K. Murray Lab – Investigates the molecular determinants of the substrate for atrial fibrillation, the most common sustained cardiac arrhythmia. Current efforts are focused on targeting reactive lipid dicarbonyls generated by oxidative stress that mediate a major component of injury in this setting.
  • D. Patrick Lab – Studies mechanisms of inflammation in autoimmune and cardiovascular diseases. Dr. Patrick has defined a unique pathway for immune activation by isolevuglandins in systemic lupus and how these contribute to hypertension in this disease. He is also investigating mechanisms of how isolevuglandins cause formation of neutrophil extracellular traps and the role of this in hypertension.
  • S. Peck Lab – Focuses on understanding the role that extracellular matrix (ECM) components play in the development and homeostasis of musculoskeletal tissues, as well as identifying mechanisms behind pathologic dysregulation of the ECM that lead to disease and degeneration. Dr. Peck's lab combines analytical chemistry, biochemistry, and molecular biology methods to identify the chemical composition of the ECM and how particular components regulate biological function of musculoskeletal cells, ultimately aiming to apply our findings to developing new regenerative therapeutic strategies for tissues such as bone, intervertebral disc and cartilage.
  • E. Rendina-Ruedy Lab – Focused on developing a comprehensive understanding of how metabolic pathways impact bone health. As such, the Rendina-Ruedy lab has ongoing projects aimed at understanding how bone cells, and cells within the bone marrow niche, store, mobilize, and utilize various metabolic substrates.
  • Roden Lab – Investigates how genomic variation affects variability in disease susceptibility and response to drug treatment. The Roden Lab is interested in the genomics of abnormal heart rhythms and use diverse tools, from examining cohorts in large biobanks to studies in wild-type or edited cardiomyocytes derived from induced pluripotent stem cells to high-throughput function in collaboration with the Glazer and Kroncke labs.
  • M. Santisteban Lab – Investigates hypertension as a risk factor for cognitive impairment, utilizing mouse models of hypertension to assess the mechanisms leading to cognitive decline. This overarching plan more specifically includes three distinct, but complimentary projects including (1) defining the trafficking and molecular mechanisms leading to immune activation in the dura (and other brain border immune populations) during hypertension, (2) profiling brain vascular and perivascular cells to reveal the molecular bases of blood-brain barrier heterogeneity and regional susceptibility to disruption during hypertension, and (3) uncover the basic mechanisms leading to hippocampal neuronal dysfunction in hypertension. The lab employs a combination of genetic, pharmacological, and advanced imaging techniques to investigate the effects of hypertension on neuronal function and cognitive health.
  • C. Shibao Lab – Focus on the pathophysiology and treatment of autonomic disorders and parasympathetic regulation of inflammation and endothelial dysfunction.
  • C. Stein Lab – Dr. Stein uses translational approaches to define the mechanisms underlying interindividual variability in drug response and toxicity. His research has defined genetic variants that underlie untoward responses to drugs and that predispose to severity of illnesses like tuberculosis, sepsis and autoimmunity.

Spotlight on the Kirabo Lab

Annet Kirabo, DVM, MSc, PhD, and her team studied the effects of salt intake on blood pressure, and found that patients who eat in excess of the American Heart Association's recommendation of one teaspoon of salt per day have altered gut microbiota, which leads to high blood pressure and inflammation.