Skip to main content

They are developing new treatments to mitigate cardiovascular pathologies induced by obesity and diabetes and unique trans-tissue drug delivery systems for localized treatments to prevent diabetic foot ulcers. Their efforts are also directed at developing a lung treatment to mitigate chronic infection and inflammation in the lower airways of the lungs via localized deep-tissue drug delivery. Past research from the Pulakat Lab identified cardiac-specific cytokine and microRNA markers associated with the progression of diabetes and in response to treatment with Rapamycin, an anti-aging drug. They also elucidated the structure-function relationship of cardiovascular reparative Angiotensin II (Ang II) receptor AT2R and novel signaling networks of the AT2R that can mitigate the progression of heart disease and vascular damage caused by diabetes and obesity.


Current projects include:

The core focus of the Pulakat lab is the elucidation and manipulation of various biological pathways that contribute to the development and pathogenesis of cardio-metabolic diseases and aging and uncovering markers of sex differences in heart disease.

The Kapur Lab was the first to establish that reduced endoglin activity improves survival and limits maladaptive cardiac remodeling in heart failure. The laboratory identified that targeting endoglin using an antibody-mediated approach limits and reverses established cardiac fibrosis in preclinical models of heart failure and acute myocardial infarction. The Kapur Lab was the first to identify that acute mechanical unloading of the left ventricle activates a cardioprotective signaling program in the setting of acute myocardial infarction and the first to identify novel molecular mechanisms regulating cardiac biology and mitochondrial function with extracorporeal membrane oxygenation (ECMO). Dr. Kapur is an inventor on multiple issued patents several of which have formed the basis for new companies.

 

Current projects include:

The Kapur Lab focuses on molecular mechanisms of cardiac remodeling and myocardial recovery. The laboratory has generated novel insight into signaling via the transforming growth factor beta (TGFb) family with a particular focus on the TGFb co-receptor endoglin (CD105) and Bone Morphogenetic Protein 9 (BMP9). The lab’s translational research focuses on preclinical models of acute and chronic heart failure, invasive hemodynamics, circulatory support device development, and cardioprotective mechanisms in the setting of acute myocardial infarction.
jaffe lab iris sitting at desk

A major area of interest is the role of the hormone aldosterone and the receptor by which it functions, the mineralocorticoid receptor (MR), in the molecular mechanisms of cardiovascular disease. The steroid hormone aldosterone is the final step in the renin-angiotensin-aldosterone pathway that regulates blood pressure and electrolyte homeostasis by activating MR in the kidney to regulate genes involved in renal sodium handling. We and others have demonstrated that MR is expressed in cells of the human blood vessel, supporting the possibility that direct effects of aldosterone on the vasculature could play a role in vascular function and disease. This is clinically significant because, in human clinical trials, pharmacologic inhibition of the MR prevents heart attacks, strokes and cardiovascular deaths with minimal effects on systemic blood pressure.

A second major area fits in the field of cardio-oncology. The Jaffe lab is exploring the impact of cancer treatment on blood vessels and how this might contribute to side effects in cancer survivors. We are exploring the molecular mechanisms for these clinical findings in order to gain a better understanding of the mechanisms of cardiovascular disease and to identify novel therapeutic drug targets for common vascular diseases or to protect cancer patients from adverse side effects.

The Jaffe laboratory routinely uses in vitro molecular and cellular biology methods, primary human vascular cell culture models, state-of-the-art transgenic mouse models with in vivo capabilities to study rodent models of cardiovascular physiology and disease, unbiased transcriptomics, proteomics, and epi-genomics methods, and translational studies utilizing human samples from our patients across Tufts Medicine.

Current projects include:

The Jaffe laboratory is focused on understanding the molecular mechanisms by which blood vessels become dysfunctional to lead to common cardiovascular conditions, including heart attack, stroke, high blood pressure, in-stent restenosis, vein graft failure and heart failure. We are interested in understanding how traditional risk factors like aging, obesity, high blood pressure or high cholesterol, or new risk factors like novel cancer treatments, cause blood vessels to become diseased. We are also focused on understanding sex differences in how these cardiovascular diseases develop, in order to identify sex-specific precision medicine strategies.
Icli Lab focuses on the role of microRNAs, a class of non-coding RNAs, in ischemic vascular disease states such as myocardial infarction, diabetic wound healing and obesity-induced chronic inflammation. Our group studies these complex disease states using diverse model systems including human plasma and tissue samples, human organoids, transgenic mouse models as well as primary cells. We take a multifaceted approach to delineate the disease mechanisms by utilizing state-of-the-art molecular biology techniques, bioinformatic approaches, bulk and single-cell RNA-seq, immunohistochemistry, and live animal imaging.

Current projects include:

The overarching goal of the lab is to identify genes associated with human heart disease. We believe firmly in the benefit of making primary discoveries advancing understanding of human disease whenever possible, in humans. Within the past two decades, the human genome sequence has been completed and a map of genetic markers suitable for studying disease associations has been established. Both candidate gene and genome-wide association studies are performed in the Huggins laboratory with a focus on valve disease and genetic cardiomyopathy.

The Good Lab takes an interdisciplinary approach encompassing fundamental molecular and cellular biology, in vivo state-of-the-art imaging technologies, ex vivo vascular function techniques, and in vivo pathophysiological models to further our understanding of how blood flow regulation and inflammation contribute to the etiology of neurological diseases and to dissect the role of arterial and venous endothelial Panx1 in ischemic stroke and Alzheimer’s Disease.

Current projects include:

The Good Laboratory is focused on understanding the mechanisms that regulate blood vessel function and inflammation and their contribution to the development of neurological diseases. Alzheimer’s Disease (AD) and ischemic stroke are both in the top 10 leading causes of mortality in the US, however, therapeutics are severely lacking to improve outcomes in these patients. CBF dysregulation, blood-brain barrier (BBB) dysfunction and neuroinflammation are key features observed chronically in Alzheimer’s Disease patients and acutely in ischemic stroke patients. A gap in our knowledge, and an area with high potential for pharmacological intervention, is understanding vascular-mediated mechanisms that influence ischemic stroke outcome, such as recovery of cerebral blood flow (CBF) and inflammatory responses, and progression of Alzheimer’s disease.

Using whole animal physiology, in vivo electrical stimulation of the heart and echocardiography, we study the predisposition of the heart to develop lethal arrhythmias and its association with diastolic dysfunction (heart failure with preserved ejection function, HFpEF). On the cellular level, we use isolated cardiomyocytes from these animal models, for cellular electrophysiology studies and direct measurements of calcium transients to measure abnormalities of repolarization, dysfunction and distribution of ion channels, and calcium dyshomeostasis in the predisposition to the development of ventricular arrhythmias. Using molecular biological tools, we study the role of second messenger signaling in the regulation of ion channel activity in the predisposition to ventricular arrhythmias, and development of diastolic dysfunction. We also have an interest in the role of Toll-Like receptors in the pathogenesis and treatment of mouse models for abdominal aortic aneurysms.

Current projects include:

The Galper Lab has a long-standing interest in autonomic control of the heart and more recently in the pathogenesis of lethal ventricular arrhythmias following myocardial infarction in mouse models for type II diabetes and metabolic syndrome. We also have an interest in the mechanism of atrial fibrillation in these mouse models for metabolic heart disease.

Cardiovascular disease (CVD) remains the leading cause of death around the world and its incidence increases greatly with age in both males and females. However, the rate of CVD development increases significantly more in postmenopausal females. Our efforts are focused on identifying alternative strategies outside of hormone replacement therapy to mitigate CVD risk in aging women, and potentially men, as our approaches allow for the direct comparison of males and females and potential therapeutic targets that we identify may also aid in male CVD.

For these studies, we routinely utilize our state-of-the-art transgenic mouse models, in vitro molecular and cellular biology techniques, ex-vivo resistance vessel reactivity via wire myography, in vivo telemetric blood pressure measurements and in vivo cardiac and vascular imaging. Additionally, our laboratory also performs clinical studies examining vascular function in several populations. For these studies, we measure 1) microvascular function in the cutaneous circulation via laser Doppler flowmetry coupled with intradermal microdialysis and 2) arterial stiffness via applanation tonometry. Thus, we are a truly translational laboratory with studies spanning from the bench to the mouse to the human.

Current projects include:

The effects of sex hormones and their receptors on vascular function in health and disease states.

Isabelle Reveillaud-Draper earned her PhD. in Biochemistry from the Université Montpellier 2/ Sciences et Techniques, Montpellier, France, studying the structural organization of small nuclear ribonucleoproteins. She then joined Dr. Tom Kornberg's laboratory at the University of California, San Francisco, for a post-doctoral position working on Drosophila development. As a research associate at the Linus Pauling Institute of Science and Medicine in Palo Alto, CA, she pioneered the expression of mammalian genes in transgenic Drosophila (i.e., mutant flies expressing the bovine superoxide dismutase) to facilitate investigations of the free radical theory of aging.

The Chin lab is interested in the molecular mechanisms that mediate the development of cardiomyopathies, with a particular focus on the genetic events that occur within the myocardium that result in myocardial dysfunction. Major areas of interest include the pathogenesis of hypertrophic cardiomyopathy (HCM), an inherited disorder that affects approximately 1 in 500, through transcriptomic, proteomic and metabolomic assessment, and the development of an enzyme replacement therapy for Barth Syndrome (BTHS), an inherited cardiomyopathy resulting from mitochondrial dysfunction.
Jump back to top