For the latest COVID-19 campus news and resources, visit umassmed.edu/coronavirus.

Search Close Search
Search Close Search
Science-related icons arranged in a design
Page Menu

Interdisciplinary Graduate Program

Many complex biological and biomedical problems are best analyzed through an interdisciplinary combination of approaches that may include classical genetics, molecular genetics, genomics/proteomics, biochemistry, and light and electron microscopy. The Interdisciplinary Graduate Program (IGP) supports and encourages graduate student training in interdisciplinary approaches to biomedical research. The IGP is characterized by a flexible graduate curriculum that is tailored to the specific needs of individual students and encourages rapid initiation of full time thesis research. The 141 distinguished faculty that participate in the program are affiliated with 16 basic science and clinical departments at the Medical School.

REQUIREMENTS FOR SPECIALIZATION

All Basic Biomedical Science students must complete the core curriculum as well as electives required by their program. IGP does not require specific advanced courses; rather, advanced coursework, journal clubs, and other enrichment activities that can be selected from those offered by any Morningside Graduate School of Biomedical Sciences program. Selections are tailored to the requirements of each student and are determined after discussion between the faculty advisor and  the student. In general, a minimum of three graded elective courses of 2-4 credits each are required. As part of the students' training, they are also required to present their research data once a year in a seminar series designed to give students an opportunity to learn scientific information and presentation skills.

View PhD Program Schedule  |  View Courses

OUR LEADERSHIP & FACULY

PROGRAM DIRECTORS

Craig Peterson, PhD
Professor
email Dr. Peterson
Visit the Peterson lab site
  Heidi Tissenbaum, PhD
Professor
email Dr. Tissenbaum
Visit the Tissenbaum lab site

IGP Dirs.jpg

OUR FACULTY

Inherent in the philosophy of the IGP, as well as the fact that the majority of GSBS faculty are members of the IGP, our faculty are involved in research areas that span the breadth of biomedical research. Consequently, studies on biochemical mechanism, molecular structure, virology, immunology, cancer biology, and neurobiology, are among their research areas.

View the affiliated faculty listing for the Interdisciplinary Graduate Program.

OUR STUDENTS

STUDENT EXPERIENCE

Numerous IGP students have been honored with a Weintraub award; a nationwide award given to less than 15 students each year based on the quality, originality, and significance of their research.  The Weintraub award has been called the "Nobel" of thesis awards by Nobel Laureate and IGP faulty member Dr. Craig Mello. Previous IGP award winners include Alla Grishok (2002), Pedro Batista (2010)  Emma Watson (2014), and Colin Conine (2014).

The IGP program sponsors a weekly seminar program where students present their research to a diverse audience of students, postdocs, and faculty.

OUR STUDENTS IN THE NEWS

Getting Results…
  • PhD student Megan Honeywell receives prestigious Ruth L. Kirschstein Predoctoral award
    Research News

    PhD student Megan Honeywell receives prestigious Ruth L. Kirschstein Predoctoral award

    In her preliminary studies, PhD student Megan Honeywell has found that cells can trigger a cell death even when they do not have the p53 protein, the so-called “guardian of the genome.”

    Read more
  • Biomedical sciences student works toward a future in genetic engineering
    Education News, Media

    Biomedical sciences student works toward a future in genetic engineering

    PhD student Katya Makeyeva investigates how small RNA pathways regulate gene expression of reproductive cells in the lab of Nobel Laureate Craig Mello, PhD.

    Read more
  • Scientific discovery propelled Cesar Bautista Sotelo on journey from Texas to Massachusetts
    Education News

    Scientific discovery propelled Cesar Bautista Sotelo on journey from Texas to Massachusetts

    Cesar Bautista Sotelo, PhD candidate in the Graduate School of Biomedical Sciences, is using cutting-edge techniques to study how cells and the genome shed offer information on human development, aging and disease.

    Read more
  • PhD candidate Mohona Gupta seeks answers in retinal degenerative disease
    Education News, Media

    PhD candidate Mohona Gupta seeks answers in retinal degenerative disease

    Mohona Gupta, third-year PhD candidate in the Graduate School of Biomedical Sciences, moved to Massachusetts from Kolkata, India in 2018 to attend UMass Medical School. It was the first time she had ever left her hometown, her family and her house for more than a few days.

    Read more
  • MD/PhD candidate Rachel Stamateris focused on novel diabetes approach
    Media, Research News

    MD/PhD candidate Rachel Stamateris focused on novel diabetes approach

    As the diabetes epidemic continues to grow, researchers are redoubling their efforts to understand the mechanisms that cause the disease. Learn about how MD/PhD candidate Rachel Stamateris is studying the disease in this Women in Science video.

    Read more

EXTERNAL AWARDS FOR RESEARCH TRAINING (CURRENT)

  • Larsen.jpg

    Investigating Trisomy 21 Impact on Human Neural Cell Development and Function Using "Trisomy Silencing" in vitro

    Trisomy 21 is thought to broadly impact the development, transcriptome, and activity of many neural cell types. However, the extent to which DS alters each is conflicted and potentially overestimated by variability not due to trisomy. Reducing variability through “trisomy silencing” will increase the power to detect legitimate effects of trisomy 21 in different neural cell types, when during development changes occur, and which remain correctable.

    Read more
  • Luu.JPG

    Investigating the role of MS4As during Alzheimer's disease

    Alzheimer’s Disease (AD) is the most common neurodegenerative disease in the world and the 6th leading cause of death in the United States. Despite significant effort, current AD therapies are highly limited in both number and efficacy. Hope for new therapeutic interventions has emerged from recent genome-wide association studies (GWAS), which have identified genes whose mutations are linked to altered AD susceptibility. One of the strongest and most reproducible genetic associations with altered AD risk are members of the Membrane Spanning 4a (MS4A) gene family. In fact, current genetic data suggest that MS4A polymorphisms account for approximately 10% of all AD cases. However, a limited understanding of MS4As has hindered the development of therapies targeting these proteins. Single cell transcriptional profiling has revealed that the Ms4a genes genetically linked to AD are selectively expressed in a subset of microglia, the resident innate immune cells of the central nervous system. Furthermore, MS4A-positive subsets of microglia display a phenotype similar to microglia seen in neurodegenerative disease states, necessitating inquiry into the role of MS4As in microglia. We found that Ms4a-positive microglia are enriched for phagocytic machinery versus Ms4a-negative microglia, and animals deficient for individual Ms4a family members have significantly reduced expression of genes important for phagocytosis compared to wild-type (WT) animals. Microglial phagocytosis is a dynamic process by which brain debris, dying cells, unwanted synaptic connections, and toxic molecules are eliminated and disruption of this process is thought to underlie numerous neurological disorders, including AD. Thus, this proposal will test the hypothesis that MS4As regulate microglial phagocytosis and alter AD pathogenesis. To test this hypothesis, aim 1 will examine microglia phagocytosis of synapses, dead neurons, and amyloid-beta 42- one of the neurotoxic, pathogenic agents of AD- both in vitro and in vivo using Ms4a-deficient and WT microglia. These experiments will characterize the role of MS4As in microglia, and provide insight into how MS4As affect microglial phagocytosis. Aim 2 will investigate the role of MS4As in AD pathogenesis. Although GWAS studies have strongly linked MS4A polymorphisms to AD susceptibility, the role of MS4A genes in AD is unknown. Some MS4A alleles confer AD protection while others increase AD susceptibility, and MA4A polymorphisms are often located in non-coding regions of these genes. To investigate whether MS4As are protective against or contributive toward AD, mice that are homozygous deficient for individual Ms4a family members will be crossed to the 5XFAD mouse model of AD and pathological features of AD, including behavioral defects in memory, amyloid beta plaque formation, microgliosis, neuronal loss, and synapse elimination will be assessed. Together, these aims will provide fundamental new insight into the role of MS4As in AD and contribute to the development of therapeutic approaches targeting MS4A receptors.

    Read more
  • Hildebrand_S.jpg

    The Impact of Nucleotide Modification Patterns on Therapeutic Small Interfering RNA Activity in the Central Nervous System

    Huntington’s Disease (HD) is caused by a CAG trinucleotide repeat expansion in exon 1 of the Huntingtin (HTT) gene that produces mutant HTT mRNA and protein. Expression of mutant HTT leads to progressive neurodegeneration via mechanisms that are poorly understood. Because HD is a genetically-defined disease, it is an ideal candidate for study and therapeutic intervention by small interfering RNAs (siRNAs) – which incorporate into the RNA-induced silencing complex (RISC) to target and degrade disease-causing genes. With the development of a divalent (di)-siRNA chemical architecture, siRNAs can be delivered throughout the central nervous system (CNS) of rodents and non-human primates. To ensure stability in the CNS, di-siRNAs require chemical modifications on every nucleotide. However, chemical modifications can affect siRNA activity and cellular localization – limiting the utility and flexibility of di-siRNAs in the CNS. The most common nucleotide modifications in siRNA replace the 2′OH of the ribose with 2′-Fluoro (2′F) or 2′-O- Methyl (2′OMe). Recent work suggests that incorporation of 2′OMe and 2′F at certain nucleotide positions may hinder siRNA loading into RISC or target binding/cleavage by RISC, and may alter the nuclear-to-cytoplasmic localization of siRNAs. Yet, the limited scope of this work has made it difficult to identify general design parameters for efficacious, compartment-specific siRNA. With guidance from Drs. Anastasia Khvorova (siRNA chemistry), Neil Aronin (HD), Phillip Zamore (RNA biochemistry) and Athma Pai (RNA sequencing), this proposal will systematically assess the impact of modification patterns on siRNA efficacy and cellular localization in the CNS to optimize siRNAs as an HD therapy and research tool for dissecting HD pathology. Aim 1 will characterize how siRNA chemical modifications impact RISC loading in vivo and target binding and cleavage in vitro. To measure how modifications alter RISC loading, a pool of differentially-modified siRNAs will be injected into the CNS of mice, RISC will be pulled down and loaded siRNAs will be sequenced. To determine the effect of siRNA chemical modifications on RISC-target interactions, target binding and cleavage kinetics will be measured for a panel of modified siRNAs using single-molecule total internal reflection fluorescence microscopy. Mechanistic insight into how modifications impact siRNA efficacy in the CNS will provide a framework with which to design optimized siRNAs to treat HD and other CNS diseases. Aim 2 will use the same modified siRNA pool from Aim 1 to identify optimal modification patterns for enhanced nuclear localization of siRNA in the CNS. These data will be used to design efficacious chemically-modified di-siRNAs targeting nuclear or cytoplasmic-only HTT RNA. These di-siRNA will then be injected into YAC128 HD mice and the effect on motor deficits, neurodegeneration, and striatal mRNA expression will be assessed. These results will provide valuable insight into the biology of HD and determine the potential of nuclear RNA-targeting siRNAs as a therapeutic paradigm for repeat expansion disorders with underlying RNA toxicity.

    Read more
  • Wortman.jpg

    Characterization of MS4A Chemoreceptive Function

    As animals explore their environment, they encounter millions of chemical signals that must be correctly interpreted to identify food and mates and to avoid predation. In mammals, the majority of this chemical information is detected by the sensory neurons of the olfactory system, (OSNs), which express more than one thousand distinct olfactory receptors (ORs), specialized chemoreceptors that sense volatile, chemical odorants. While many of these receptors have been molecularly defined, how they function to elicit appropriate behavioral responses remains largely unclear (i.e., how does a mouse know to run away from a cat and toward a piece of cheese). This proposal outlines a series of experiments aimed at taking advantage of the opportunities offered by the olfactory system to begin to characterize the mechanisms by which chemosensory information is detected and processed to generate specific, stereotyped behavioral responses. We have recently identified a new family of olfactory receptors encoded by the Ms4a family of genes, which our preliminary experiments suggest are required to mediate innate avoidance responses to stimuli such as predator-derived odorants that signify danger to the animal. However, how MS4A receptors function to elicit these behaviors is largely unknown, which significantly impairs our understanding of how the olfactory system interprets relevant sensory stimuli to elicit appropriate behavioral responses. To begin to elucidate MS4A chemoreceptor function, in Aim 1 I will use mutagenesis, electrophysiology, and calcium imaging to determine how MS4As bind ligands and signal their presence. In Aim 2, I will use ex vivo and in vivo preparations as well as mouse behavioral assays to determine the contribution of MS4As to transducing predator odor presence into appropriate innate avoidance responses. Together, the experiments proposed here will characterize a novel family of odorant receptor and how it signals the presence of ethologically relevant odors. Broadly speaking, these studies seek to illuminate basic principles of olfaction, provide insight into mammalian chemosensation, and shed light on the neural pathways associated with sensory perception.

    Read more
  • Ferguson.jpg

    Tissue-specific modulation of Apolipoprotein E in neurodegeneration

    Alzheimer’s Disease (AD) and Amyotrophic Lateral Sclerosis (ALS) are multifaceted, progressive neurodegenerative conditions that place a monumental burden on patients, providers, and the public healthcare system. No disease-modifying treatments are currently available for either AD or ALS. Although the etiology of each disease is well studied, strategies targeting characteristic features of disease pathogenesis— e.g., beta-amyloid in AD—show limited clinical efficacy. Identification of novel targets that modify progression of neurodegeneration is needed for innovative therapeutic development across neurodegenerative disorders. AD and ALS are caused by genetic and environmental factors that alter downstream pathways like lipid homeostasis. A critical player in systemic and central nervous system (CNS) lipid transport, that is also implicated in the onset and progression of neurodegeneration, is <em>apolipoproteinem> E (ApoE). Genetic deletion of ApoE reduces neuropathology in mice, but also causes atherosclerosis. Thus, despite its implication in disease, the diverse functionality of ApoE in its distinct biological “pools” (i.e. systemic and CNS) makes it a challenging therapeutic target. Reducing individual ApoE pools may circumvent this issue. However, the independent effects of systemic or CNS ApoE silencing on neurodegenerative diseases are unclear. The goal of this proposal is to determine the relationship between systemic and CNS ApoE pools, and their effects on disease progression in AD and ALS mice. The project will take advantage of chemically-stable, self- delivering small interfering RNAs (siRNAs) that enable sustained, <em>tissue-specificem> silencing of target mRNA. GalNAc-siRNAs specifically deliver to liver (site of systemic ApoE production), and divalent (Di)-siRNAs deliver throughout the brain and spinal cord after intra-cerebroventricular (ICV) injection. With guidance from Drs. Anastasia Khvorova (siRNA chemistry), Robert Brown (ALS), Evgeny Rogaev (AD models), Andrew Tapper (animal behavior), and Thomas Smith (neuropathologist), GalNAc- and Di-siRNA will be used to silence hepatic and CNS ApoE, respectively, and the effects on CNS and systemic ApoE pools, and AD and ALS phenotypes, will be examined. In Aim 1, GalNAc-siRNA targeting ApoE will be subcutaneously injected into mice. In Aim 2, Di-siRNA targeting ApoE will be delivered to the CNS via ICV injection. Both aims will utilize the APP/PSEN1 mouse model of AD and the SOD1G93A mouse model of ALS, and will measure systemic and CNS ApoE and cholesterol levels, and AD and ALS neuropathology and behavior two months post injection. These studies will advance the understanding of how ApoE pools interact in the context of neurodegeneration, and the effects on disease progression. Such findings will inform strategies for safe and effective therapeutic targeting of ApoE in AD, ALS, and age-related neurodegenerative disorders.

    Read more
  • Bamidele.jpg

    Engineering Synthetic Guide RNAs and Compact Base Editors for Enhanced In Vivo Delivery

    CRISPR-Cas9 technology has profoundly advanced genetic research and gene therapy by enabling rapid, precise, and programmable genome editing to study gene function or correct mutations in cells and in vivo. Among CRISPR-mediated gene editing approaches, cytidine and adenine base editors (CBEs and ABEs) enable efficient and precise single-base transitions in dividing and non-dividing cell types. Base editors comprise a catalytically-impaired nickase Cas9 (nCas9) fused to a cytosine deaminase (CBE) or adenine deaminase (ABE) that is guided to a target site by a “guide RNA”. With the potential to enable all four nucleotide transitions in the context of a base-pair (C:G→T:A or A:T→G:C), base editors have the potential to cure a wide range of genetic disorders. Realizing this hope requires efficient and safe in vivo delivery methods. In vivo delivery of base editors relies on adeno-associated virus (AAV) vectors. Due to the limited packaging capacity of AAVs and the large size of nCas9 orthologs (e.g., SpyCas9 from Streptococcus pyogenes), delivery requires two AAVs encoding an intein-split nCas9-BE that fuses into a functional complex when co-expressed in cells. Dual AAVs encoding intein-split SpyCas9-BEs achieve therapeutically-relevant levels of base editing in pre-clinical disease models, including in the central nervous system (CNS). Nonetheless, dual AAV SpyCas9-BE delivery suffers from several limitations, including: toxicity from increased viral load; high vector production costs; immune response and off-target editing caused by sustained expression of nCas9-BE components; and limited ability of guide RNA to specify multiplexed edits. Under guidance from Drs. Erik Sontheimer (CRISPR), Miguel Sena Esteves (AAV delivery), Anastasia Khvorova (oligonucleotide chemistry), and Athma Pai (sequencing, bioinformatics), this proposal aims to develop flexible delivery approaches for efficient and safe base editing in vivo. This project will take advantage of established gene therapy modalities, including a single AAV vector encoding a compact Cas9 from N. meningitidis (Nme2Cas9), and chemically-modified oligonucleotides. Aim 1 will optimize and validate an all-in-one AAV encoding a compact Nme2Cas9-ABE and its guide RNA for efficient in vivo base editing in mice. The use of a compact ABE for AAV delivery will decrease viral load, production costs and may increase delivery efficiency. Aim 2 will develop chemically-modified Nme2Cas9 crRNA (target-specify portion of guide RNA) for co-delivery separate from an AAV encoding Nme2Cas9-ABE and tracrRNA (invariant portion of guide RNA). This approach will provide a way to control nCas9-BE expression and streamline multiplexed base editing via delivery of multiple crRNA. Although these delivery approaches are applicable to variety of tissues, this study will focus on the CNS, where AAV- and oligonucleotide-based therapies have shown some success, but a dire need for transformative therapeutics remains. Completion of this study will establish novel delivery approaches to advance the utility of CRISPR for in vivo applications, including functional genomic studies and base editing therapies.

    Read more
  • Durning.jpg

    Investigating how the conserved ZNFX-1 protein regulates epigenetic inheritance and germline immortality in Caenorhabditis elegans

    Germ cell immortality is essential for fertility and for species survival. To proliferate indefinitely, germ cells depend on mechanisms that maintain genome integrity and epigenetic programs. In animals, small RNAs that interact with Argonaute proteins of the PIWI family—called piRNAs—serve as a vanguard of transcriptome and epigenome integrity in the germline, by identifying and silencing transposable elements and by regulating germline gene expression. In many animals, piRNAs are essential for germline health. Defective piRNA biogenesis or function activates transposon expression and mobility, increases DNA damage, disrupts germ cell development, and reduces fertility. In Caenorhabditis elegans, small RNA pathways with opposing activities collaborate to maintain germ cell survival and fertility. Recent studies identified ZNFX-1 as a regulator of epigenetic inheritance in worms. ZNFX- 1 is a highly conserved UPF1-like helicase with C-terminal NF-X1-type zinc finger domains. znfx-1 mutants activate epigenetically silenced reporters, and in some cases they silence normally active reporters, indicating that ZNFX-1 balances opposing epigenetic programs. The targeting pattern of small RNAs redistributes in znfx- 1 mutants, suggesting that ZNFX-1 determines the origin of small RNAs required for silencing and anti-silencing pathways. Preliminary data also show that znfx-1 null mutations cause a mortal germline phenotype at elevated temperatures, suggesting that ZNFX-1 maintains balanced epigenetic signals essential for germline immortality. This proposal seeks to use genetic, computational, and biochemical approaches to test the hypothesis that ZNFX-1 is recruited to targets and identifies sites of small RNA biogenesis. Studies in Aim 1 will determine how ZNFX-1 regulates small RNA biogenesis by identifying how and where it binds to target transcripts, and if it unwinds or moves along RNA. Studies in Aim 2 will determine how ZNFX-1 regulates germline immortality by identifying functional domains of ZNFX-1, potentially redundant proteins with homologous domains, and small RNA and transcriptome features of germline immortality. These studies will reveal how animals transmit heritable epigenetic information and how epigenetic pathways maintain germ cell immortality. In addition, the proposed research will provide training in genetics and epigenetics, quantitative biochemistry, and computational approaches, and prepare the fellow for a postdoc in computational and systems biology and a future career as an independent investigator.

    Read more
  • Krykbaeva.jpg

    Characterizing effects of sperm- and oocyte-derived epigenetic factors on early embryonic gene expression and offspring metabolic function

    Metabolic diseases such as obesity have become significant health risks affecting one-third of the population worldwide and can have devastating complications. It is therefore imperative to understand the causes of metabolic disease predisposition in order to develop preventative strategies. Extensive genetic studies have failed to explain this ongoing epidemic. However, parents pass on not only genetic information, but also epigenetic factors, which can be modified in response to environmental stimuli, and can then affect gene expression. If information about parental environment can be recorded in the germline, it has the potential to be transmitted to the zygote and impact offspring health. This concept remains controversial in mammals and represents a large knowledge gap in the field of embryology. Previous work in the lab has demonstrated that sperm from fathers fed a low protein diet carry tRNA fragments and microRNAs that can modify gene expression in the embryo. Therefore, in Aim 1, sperm-derived RNAs will be purified and microinjected into parthenogenetically-activated oocytes, or parthenotes, which lack any paternal genetic content. Transcriptomic profiles of injected parthenotes will be acquired by single-embryo RNA-Seq to characterize resulting alterations to the embryonic transcriptome. Maternal transmission of epigenetic information has not been characterized to the same extent as the paternal side. Therefore, in Aim 2 a similar dietary paradigm will be utilized to address the question of whether information about maternal diet can be carried in oocytes and result in changes to the embryonic transcriptome. In vitro-fertilized embryos from mothers fed low protein, high fat, or control diet will be sequenced by single-embryo RNA-Seq. These embryos will be transferred to foster mothers to produce adult offspring, which will then be assessed for glucose tolerance and insulin resistance. The use of in vitro fertilization in this paradigm will ensure that any changes observed in the embryo originate from the oocyte and not from nutrient exchange during gestation. Completion of this research will elucidate effects of paternal and maternal epigenetic factors carried by gametes on the embryonic transcriptome. This work will be completed at the University of Massachusetts Medical School under the sponsorship of Dr. Oliver Rando. The fellowship training plan includes training in embryological techniques, such as microinjection and immunofluorescent staining of lineage markers, as well as metabolic phenotyping of adult mice. Opportunities to gain experience in science communication include participation in departmental seminars and local and national conferences. Furthermore, career development workshops are provided by the university, in addition to teaching and mentoring of first-year graduate students.

    Read more
  • Denis Lafontain, Dekker Lab, Funding provided by National Institutes of Health

    Stability of the folded genome

    Perturbations in normal gene expression arising from defects in genome organization can lead to cellular dysfunctions linked to aging and various disease states. The mammalian genome is generally organized into chromosomes, compartments, topological associating domains (TADs) and loops. Although TAD and loop formation have been extensively studied, little is known about the processes that drive nuclear compartment formation. It has been proposed that microphase phase separation drives the association of genomic domains of similar chromatin state, resulting in the formation of either type A (active chromatin) or B (inactive chromatin) compartments. However, identifying factors involved has been limited by a lack of tools capable of quantifying the biophysical properties driving this phenomenon. Mammalian heterochromatin protein 1 (HP1) α and HP1β bind constitutive heterochromatin and are known to facilitate the bridging of nucleosomes, suggesting that these proteins play a key role in heterochromatin compartmentalization. Although a recent study has demonstrated that heterochromatin compaction is independent of HP1α, work from our collaborators suggest that this protein is required to stabilize interactions between heterochromatic loci. Interestingly, HP1 proteins and several of their interacting partners can bind RNAs. Independent of HP1 function, specific RNA transcripts are known to play important roles in the formation and maintenance of spatial genome organization and perhaps microphase separation, notably at nucleoli, speckles, and the inactive X chromosome of female cells. We recently developed liquid chromatin Hi-C (LC-Hi-C), which allows quantification of chromatin interaction stability measurements genome-wide. Briefly, isolated nuclei are subject to in situ restriction digestion. Digestion of the genome into a specific fragment size distribution results in the loss of low density/unstable interactions whereas higher density/stable interactions are maintained, which is quantifiable by genome-wide chromosome conformation capture (Hi-C). This technique reveals that the dissolution kinetics of chromatin interactions vary widely between A and B compartments as well as compartmental substructures. The development of “in situ LC-HiC” in Aim 1 will allow stability measurement on mitotic chromosomes, streamline the existing protocol and allow the study of smaller cell populations. Aim 2 will assess contributions of (HP1) α and HP1β to stability of heterochromatic interactions. In Aim 3, LC-Hi-C will allow identification of genomic regions destabilized by RNA depletion. Candidate factors contributing to stability will then be identified using in situ chromatin-associated RNA sequencing (iMARGI) and validated by perturbation followed by LC-Hi- C. Taken together, this study aims to measure the dynamics of chromatin interactions and to provide new mechanistic insight as to how the genome is organized throughout the cell cycle.

    Read more
  • Galan.jpg

    Replication-independent DNA methylation dynamics during post-testicular sperm maturation

    It has become apparent that sperm are extensively remodeled during epididymal transit, with ongoing changes to the protein and RNA content of maturing sperm. In addition, an increasing number of studies have shown that epigenetic information is modified during the process of sperm maturation. During the first year of my PhD studies, I have discovered that cytosine methylation patterns are surprisingly dynamic during sperm maturation in the epididymis. Using whole genome bisulfite sequencing (WGBS), I identified widespread changes in DNA methylation as sperm move from the testes through the caput, corpus, and cauda of the epididymis. Given that sperm are a highly methylated and condensed cell type, it is surprising and novel that epididymal transit results in any DNA methylation reprogramming of sperm. I aim to address the question of how DNA methylation changes occur between different sperm populations. My approach is to recapitulate maturation-associated cytosine methylation dynamics in vitro to understand the mechanism by which these changes are occurring. Resolving what these changes mean for potential offspring and determining the mechanisms and factors leading to changes in methylation will have significant implications for both assisted reproduction, and for understanding how a father's environment impacts his children's well being.

    Read more

EXTERNAL AWARDS FOR RESEARCH TRAINING (PAST)

  • Stamateris.jpg

    Mechanism of cdk4 diabetes rescue in IRS2 knockout mice

    Type 2 Diabetes (T2D) is a major public health issue in the United States with approximately 9.3% of the population suffering from the disease. Additionally, 86 million people have prediabetes and the economic impact is staggering, with 1 in 10 health care dollars being spent on T2D and its complications. T2D results from insulin resistance and reduced beta cell mass; thus, strategies to increase functional beta cell mass are critical goals for diabetes research. Although it is well established (from rodent models) that increased beta cell mass results from enhanced beta cell proliferation, new research suggests that beta cell dedifferentiation also contributes to reduced beta cell function. Some proteins involved in the G1/S transition of the cell cycle, especially Cdk4, are critical for the maintenance of beta cell proliferation and mass. Insulin receptor substrate 2 knockout (Irs2 KO) mice develop diabetes due to peripheral insulin resistance and reduced beta cell mass, and we previously found that in vitro re-expression of cyclin D2, which activates Cdk4, rescues the loss of proliferation in beta cells lacking Irs2. Therefore, we hypothesized that expression of a constitutively active form of Cdk4 (Cdk4 R24C) might be able to rescue the diabetic phenotype of Irs2 KO mice. Intriguingly, preliminary results suggest that Cdk4 R24C is able to completely rescue not only beta cell mass, but also insulin secretion and beta cell differentiation. Interestingly, recent studies show that the Cdk4 kinase plays many roles independently of its known activity in the cell cycle. Therefore, the goal of this proposal is to determine the mechanisms behind this rescue and determine what atypical roles cdk4 plays in the beta cell. In Aim 1, we will determine how Cdk4 rescues beta cell proliferation, focusing on both the canonical Cdk4-Rb- E2F pathway, and will also identify novel Cdk4 interactors in the beta cell using BioID. In Aim 2, we will determine if Cdk4 R24C rescues 1st or 2nd phase insulin secretion in Irs2 KO islets using both islet perifusion and hyperglycemic clamps studies. We will also perform molecular studies to determine whether the KATPase Kir6.2, which was previously reported to be a target of the Cdk4-Rb-E2F1 pathway, is increased and is sufficient to rescue insulin secretion in Irs2 KO islets. Finally, in Aim 3 we will explore how Cdk4 R24C is able to restore beta cell differentiation markers. This is surprising and interesting, since it goes against the data showing that when beta cells proliferate they lose differentiation markers, and I think the most likely explanation is that Cdk4 is having effects unrelated to its cell cycle actions. I will investigate how Cdk4 rescues Pdx1 expression, with a focus on FoxO1 and PPARγ, two transcription factors that regulate Pdx1 expression. Using in silico analyses and reading the primary literature, I found that both contain Cdk4 consensus phosphorylate sites. Therefore, I will determine whether Cdk4 acts via either or both of these to maintain beta cell differentiation. If Cdk4 plays atypical roles as a kinase to influence multiple aspects of beta cell biology, this may lead to better therapeutic options for preserving beta cell mass, function and differentiation in T2D.

    Read more
  • Davis.jpg

    Defining the Rules for Designing Fully Chemically Modified siRNAs to Treat Genetically Linked Central Nervous System Disorders

    Small interfering RNA (siRNA) therapeutics are a promising class of drugs for the treatment of genetic disorders. Chemical modification of siRNA is necessary for delivery to the central nervous system, but may hinder the efficacy of some siRNAs. This project will define the relationships between siRNA sequence, chemical modification patterns, and efficacy to streamline the development of clinically-relevant siRNAs capable of treating genetically-defined neurological disorders.

    Read more
  • Luk.jpg

    Amelioration of Beta-hemoglobinopathies by efficient precise deletion of the +58 BCL11A enhancer using orthogonal Cas9-Cas9 chimeras

    Project Summary β-Thalassemia and sickle cell disease (SCD) are severe inherited anemias that result in defective β-globin expression. A common feature shared between both disorders is that the diseases can be mitigated by the production of fetal hemoglobin (HbF). Current treatments for both disorders are mainly supportive and focus on alleviating disease complications. However, management of these β-hemoglobinopathies is expensive, restrictive, lifelong, and associated with significant side effects. Currently, allogeneic hematopoietic stem cell therapy is the only curative treatment for β-hemoglobinopathies. However, finding human leukocyte antigen (HLA)-matched donors is highly challenging. Gene editing approaches that alter the expression of HbF should improve the health and quality of life of individuals with β-hemoglobinopathies. The goal of this project is to develop efficient, accurate, and safe CRISPR gene editing approaches as a universal treatment for β-hemoglobinopathies. Typically, a nuclease (SpyCas9) and guide RNA (gRNA) target a specific region (+58 enhancer of BCL11A) and creates a double-stranded break. Small insertions or deletions (InDels) are created when imprecise repair of the DNA break occurs, resulting in inactivation of the target gene or functional element. Current therapeutic editing approaches for β-hemoglobinopathies at the BCL11A locus focus on the mutagenesis of a single GATA1 recognition sequence within the +58 enhancer in CD34+ hematopoietic stem and progenitor cells (HSPCs), limiting the scope of their potential impact. This project will harness our recently developed Cas9-Cas9 fusion chimera, which is able to produce segmental deletions with defined junctions (precise deletions) at a higher efficiency than standard Cas9 nucleases. Furthermore, Cas9-Cas9 fusions are able to effectively target suboptimal PAM sequences, and thus these nucleases have a broader targeting range than standard SpyCas9. These properties make Cas9- Cas9 fusions ideal for the deletion of therapeutically relevant regulatory elements, such as the +58 enhancer of BCL11A. Our preliminary studies show that ribonucleoprotein (RNP) complexes of these Cas9-Cas9 fusions targeting this locus are functional in CD34+ HSPCs and result in robust induction of HbF. In Aim 1, I will define deletion products within the +58 enhancer that facilitate the maximum induction of HbF in erythroid model systems and optimize the Cas9-Cas9 fusions to efficiently and specifically produce these precise deletions with minimal collateral damage to the genome. In Aim 2, optimized Cas9-Cas9 fusion protein-sgRNA complexes will be delivered into CD34+ HSPCs and HbF induction levels will be quantified in erythroid progeny. Treated CD34+ cells will be engrafted into immunodeficient mice to measure engraftment potential and persistence of editing in long-term hematopoietic stem cells (LT-HSCs). The genome-editing tools generated from the proposed work will provide a path to improved autologous HSC therapies that will impact the lives of individuals affected by β-hemoglobinopathies.

    Read more
  • Lee.jpg

    Ethanol's Effects on Brown Adipose Development and Function

    Approximately 7.2% of adults in the United States in 2012 suffered from an Alcohol Use Disorder, as defined by the American Psychiatric Association. Alcohol is responsible for a number of severe diseases, including alcoholic liver disease, alcohol related dementia and fetal alcohol syndrome. Past research has focused on organs such as the liver, or brain, but it has become increasingly evident that alcohol has significant effects on nearly all tissue in the body. One tissue that has garnered recent attention in part due to the rapid rise in obesity, is adipose tissue. Recent studies have provided compelling evidence relating the dysfunction of adipose tissue to the progression of many diseases, including liver disease. The interaction between ethanol and adipose tissue, particularly brown adipose tissue, has been understudied. There is little known about how ethanol impacts brown adipose tissue function, and further understanding of this relationship may prove crucial in elucidating origins of systemic changes seen in chronic alcoholics. This proposal will investigate the effects of ethanol on brown adipocyte development and function, elucidate mechanisms involved, and study the consequences of these effects on metabolic homeostasis. Both an in vitro and an in vivo model will be used. An in vitro model will be used to investigate whether ethanol interferes with the development of brown adipocytes. Brown preadipocytes isolated from wild-type C57BL/6J mice will be differentiated into mature brown adipocytes with or without exposure to 100mM ethanol. RNA and protein isolates will be collected from the differentiated cells. Markers of adipocyte character and function will be evaluated by qRT-PCR and western blot. Additionally, proteins involved in the mTOR pathway will be evaluated via western blot and immunoprecipitation to elucidate mechanisms that may be involved with brown adipocyte function. A similar approach will be applied to mature brown adipocytes. These studies are the focus of specific aims 1 and 2. An in vivo model will be used to study the effects of chronic ethanol intake on brown adipose tissue function, and to relate these effects with whole body metabolism. Wild-type C57BL/6J mice will be given a chronic ethanol diet. Groups of mice will be exposed to conditions that have been shown to mediate brown adipose tissue activity, including cold habitat, beta adrenergic injection, high fat diet (al 3 increase BAT activity), and thermoneutral habitat (decrease BAT activity). Weight and temperature will be recorded throughout the study, and mice will be sacrificed for collection of tissue. Various depots of adipose tissue will be collected, along with liver and muscle. These tissues will be evaluated for both functional markers, and markers of brown/beige/white adipocyte fate. These experiments are the focus of specific aim 3.

    Read more
  • GarciaGonzalez.jpg

    C. elegans as a model for host-microbe-drug interactions

    The bacteria that live in our body (microbiota) help us metabolize different nutrients and chemicals from our diet, including the medications we take. Thus, these bacteria can influence our response to treatments, like chemotherapy, by converting drugs into more or less toxic forms. Understanding the mechanisms by which bacteria influence our response to drugs is critical to design better treatments that maximize therapeutic effects and minimize adverse effects. The nematode C. elegans and its bacterial diet provide a suitable model to explore host-microbe drug interactions because both host and microbe are amenable to high throughput drug screening and genetic screening. I propose to use the nematode C. elegans as a model to study host-microbe-drug interactions in cancer chemotherapy drugs. In Aim 1, I will test ~ 200 cancer chemotherapy drugs for developmental or fecundity phenotypes in C. elegans animals fed two different bacteria. Additionally, I will test the role of active bacterial metabolism in the observed host-microbe-drug interactions. Then, I will use genetic screening and metabolomics, in the host and in the bacteria, to characterize the mechanisms responsible for the observed drug response. In summary, this project will generate a set of host and bacterial genes that contribute to the response to cancer chemotherapeutic agents.

    Read more
  • McFleder.jpg

    The Role of 3' End Formation in Synaptic Function

    Learning and memory are dynamic processes that require alterations in the connections among neurons. Synapses, the structures through which neurons communicate with one another, undergo biochemical and morphological changes in response to neuronal activity in a process known as synaptic plasticity. Disturbed synaptic plasticity is the basis for several diseases such as dementia, schizophrenia, and Alzheimer's disease. Local translation of mRNAs in dendrites is essential for regulating certain forms of synaptic plasticity. Impairment of this process is the cause of several neuropathies such as the Fragile-X Syndrome and other disorders linked to autism. Modulation of cytoplasmic mRNA poly(A) tail length is one mechanism that controls local translation in dendrites. It is estimated that ~7% of brain RNAs are regulated by this process in response to stimulation, which underscores its importance in synaptic plasticity and therefore higher cognitive function. Because poly(A) tails lengthen as well as shorten, there are likely to be several enzymes involved in this process. Several factors responsible for poly(A) elongation have been identified, but the enzyme(s) responsible for shortening the poly(A) tail remains unknown. The proposed research seeks to identify this deadenylating enzyme, which is likely to act as a negative regulator of dendritic translation and learning and memory. The proposed research focuses on CNOT7, a major mammalian deadenylase, in the brain and seeks to test whether (1) knockdown or mutation of CNOT7 alters poly(A) tails in cultured neuronal dendrites, (2) CNOT7 controls specific mRNA deadenylation in dendrites, and (3) knockdown of CNOT7 alters various forms of synaptic plasticity. This work will further our understanding of the process of learning and memory and disorders affecting these important processes.

    Read more

POST-DEGREE CAREERS

IGP PhD graduates have gone on to pursue careers in academia, industry, teaching, science education and more.