Department of Chemistry and Chemical Biology Research Day 2024
We invite students, faculty, alumni and staff to register for the Department of Chemistry and Chemical Biology's annual Research Day.
Get together and celebrate the research accomplishments of the CCB community
Learn about emerging AI education, research, and career opportunities
Network with students, faculty, external advisors, alumni, and friends
Come join us!
Friday, May 3, 2024 - 10 am to 3 pm
University Center Complex, TechFlex Auditorium
Registration is now closed.
Agenda
10 a.m. | REGISTRATION & CHECK-IN Coffee and Breakfast provided |
10:30-11 a.m. | WELCOME Opening remarks - Dr. Woo Lee, Department of Chemistry and Chemical Biology Event Overview – Ms. Panagiota Georgoutsos, '24 BS Chemical Biology, Past President SGA External Advisory Board (EAB) – Dr. Adam Weinglass, Executive Director, Merck Vision for AI – Dr. Nariman Farvardin, President, Stevens Institute of Technology |
11- 11:45 a.m. | STIVALA LECTURE Engineered T cell Immunity: A World of Opportunities and Challenges Introduction – Dr. Jean Zu, Lore E. Feiler Dean, School of Engineering & Science Presenter – Dr. Michael Milone, Associate Professor of Pathology, University of Pennsylvania Perelman School of Medicine Abstract: Chimeric antigen receptor (CAR) T cell therapies have transformed over the last two decades from an idea on the fringe of clinical medicine to a cornerstone of therapy for multiple B-cell malignancies including leukemia, lymphoma and multiple myeloma. Despite its success, many challenges remain to this highly bespoke engineered T cell therapy especially to its applications to solid tumors. This presentation will discuss some of these challenges and novel receptor and cell engineering approaches for improving effectiveness. We will also discuss opportunities for applying engineered T cell therapy to diseases beyond cancer such as autoimmunity. |
11:45 a.m.-12 p.m. | Q&A Moderator – Ms. Felicia Spadavecchia, Ph.D. Candidate, Chemical Biology |
12-1 p.m. | PANEL – LEARNING AND APPLYING AI FOR A CAREER IN HEALTHCARE Artificial intelligence is predicted to revolutionize how we work and live. In a fireside chat format, students have the opportunity to engage a panel of leading scientists at the frontier of genomic medicine working in academia and biopharma about their thoughts on AI education, research, and career opportunities. Introduction - Dr. Kenny Wong, Teaching Associate Professor and Associate Chair for Graduate Studies (Moderator) Speakers from our External Advisory Board Dr. Pek Lum, Chief Executive Officer and Founder, Auransa Dr. Oscar Puig, Vice President, Translational Medicine and Diagnostics, Nucleai Dr. Benjamin Tycko, Co-Director, Institute for Cancer and Infectious Diseases, Hackensack Meridian Health Center for Discovery and Innovation |
1-3 p.m. | NETWORKING OVER POSTERS & LUNCH - How to network today?: Ms. Sarah Peneiras ‘26, BS Chemical Biology Maximize your networking potential by engaging with students, faculty, EAB members, guests, and poster presenters. With each interaction, collect a sticker to win exciting prizes! - Undergraduate Student Research Projects: Dr. Sesha Alluri, Senior Research Coordinator We are proud to showcase Senior Capstone Research Projects. Senior Capstone Research is the culmination of CCB Research Spine, which provides students with the knowledge, experience, and tools needed to explore transformative scientific ideas for meaningful societal impact. This year’s projects range from drug discovery using AI to cancer research to microplastics to skin care. A panel of distinguished judges will evaluate and select three best posters. - Graduate Student Research Projects: Dr. Kwahun Lee, Assistant Professor CCB’s vision is to be a community of researchers with molecular, cellular, and computational minds. To capture this vision, our Ph.D. candidates will present their dissertation research in drug discovery, cancer research, and computational chemistry. - Pedagogical Innovation Projects: Dr. Patricia Muisener, Teaching Professor and Associate Chair for Undergraduate Studies CCB’s mission is to educate students with a broad foundation in chemical and biological sciences. The undergraduate curriculum has been innovated to highlight the department’s distinguishing features: research spine, technology emphasis, and societal context. Faculty will present their pedagogical innovation in the research spine courses as well as biology and chemistry courses. |
3 p.m. | ADJOURN |
About our Speakers
Adam Weinglass, Ph.D., Executive Director, Screening & Compound Profiling, Merck & Co
Adam Weinglass has led the Screening & Compound Profiling Department since 2018. He drives the application of cutting-edge capabilities towards the identification and progression of small molecule, cyclic peptide and degrader chemical matter.
Adam has also held positions of Director Automated Ligand Identification Systems (ALIS) & High-throughput Experimentation, Director of Cellular Pharmacology, Research Fellow (Diabetes) and Senior Research Biochemist (Ion Channels) since his hire to Merck in 2004.
Prior to Merck, Adam was a Research Fellow of the Howard Hughes Medical Institute at the University of California Los Angeles, where he worked with Drs. Kaback, Faull & Whitelegge to explore the mechanism of action of molecular membrane transporters utilizing biochemical, biophysical & mass spectral approaches.
Adam’s research has resulted in over 40 publications, reviews and U.S. patents. His discovery efforts have contributed towards several clinical candidates, research collaborations and state-of-the-art technologies. Adam received a 1st class BSc in Biochemistry & Molecular Biology from the University of Leeds and a PhD from the same Department studying the over expression, purification and characterization of recombinant mammalian sugar transporters.
Michael Milone, M.D./Ph.D., ’93 BS Chemical Biology
Dr. Milone's research is focused on the development of genetically engineered T cell immunotherapies for the treatment of disease.
He is a co-inventor of tisagenlecleucel (Kymriah), the first US FDA-approved gene therapy that employs T cells genetically modified with a chimeric antigen receptor (CAR) targeting CD19 for the treatment of B-cell malignancies.
Dr. Milone is a scientific co-founder of Cabaletta Bio (NASDAQ: CABA) and Verismo Therapeutics.
Pek Lum, Ph.D., Chief Executive Officer and Founder, Auransa
Pek Lum is the co-founder and CEO of Auransa Inc., a computationally-driven pharmaceutical company. She is the chief architect of the science behind Auransa’s technology, combining her deep understanding of genomics and her vast experience in applying data science in the pharmaceutical industry.
Before founding Auransa, Pek was an early employee and vice president of Product and chief data scientist for Ayasdi, a deep tech software company, where she created early business verticals and helped grow the company.
Pek started off her career at Rosetta Inpharmatics, a pioneer in genomics and microarray technology. After Rosetta's acquisition by Merck, Pek continued to champion the integration of data science into biology, contributing to Merck’s development pipeline. Pek did her undergraduate degree as a Monbusho Scholar at Hokkaido University, Japan and received her PhD studying yeast genetics at the University of Washington, Seattle. Her work has been widely published in scientific and medical journals, such as Nature and Cell, and her research has contributed to discoveries in drug development and the understanding of complex diseases.
Oscar Puig, Ph.D., Vice President, Translational Medicine and Diagnostics, Nucleai
Oscar Puig currently holds the position of VP Translational Medicine & Diagnostics at Nucleai, an AI-powered image analysis and biomarker development company.
Prior to joining Nucleai, Oscar held leadership positions at BeiGene, Lilly, BMS, Roche, and Merck, with a focus on biomarkers and diagnostics in oncology clinical development. Oscar Puig holds a PhD in Molecular Biology from the University of Valencia, Spain, and postdoctoral stages at EMBL, Germany and UC Berkeley, US.
Benjamin Tycko, M.D./Ph.D., Co-Director, Institute for Cancer and Infectious Diseases, Hackensack Meridian Health Center for Discovery and Innovation
Dr. Benjamin Tycko joined the recently established HMH Center for Discovery & Innovation (CDI), in Nutley NJ, as a founding member in 2018 - moving there after three decades as a faculty member at Columbia.
Starting with pioneering early studies of genomic imprinting in human development and cancer, his lab has formed a focus on genetic-epigenetic interactions, including high-resolution mapping of allele-specific DNA methylation (ASM), followed by functional validations using CRISPR-Cas9, to precisely pinpoint genetic variants that underlie inter-individual differences in susceptibility to a broad range of common human diseases.
Dr. Tycko’s lab has been continuously NIH-funded throughout his career, with his most recent grants, from the NIH and DOD, dealing with genetic-epigenetic mapping and functional assays in multiple myeloma and kidney cancers. At the same time, in collaboration with Dr. Eugene Yu at Roswell Park, his lab is elucidating an important trans-acting genetic-epigenetic interaction, in which a simple chromosomal aneuploidy produces altered DNA methylation patterns across the entire genome. This phenomenon is exemplified by human Down syndrome (trisomy 21), and the mechanistic findings in that syndrome have implications for cancers with other chromosomal and sub-chromosomal aneuploidies.
The long-term goal of all these studies, which have been published in Nature Genetics, the American Journal of Human Genetics, Genome Biology, and other journals, is discovery of genes, genetic variants, and biological pathways that can be targeted for disease treatment and prevention.
Senior Research Projects
Project name
Advances In Immunotherapy for Colon Cancer
Description
This research was an examination of the major gaps in immunotherapy for colon cancer. Current immunotherapy for colon cancer is not sufficiently adequate to resolve tumors, thus this research identified how immune cells are altered in colon cancers. The impact of immune cells such as macrophages, B cells, T cells, MDSCs, etc. were studied in the canonical Wnt pathway and the Notch signaling pathway, crucial pathways to the development and metastasis of colon cancer. Genes and proteins like SPDEF and GALNT7 were studied to determine their role in the Notch pathway, and their collaborative impact on the Notch pathway. Both SPDEF and GALNT7 were examined for their role in stemness and the progression of colon cancer. From the literature review, it was determined that SPDEF functions as a tumor suppressor in colon cancer via the suppression of the Wnt genes through β-catenin, and GALNT7 does not have a direct relationship with colon cancer. Ultimately, the goal of this research is to use the literature reviewed to improve research into immunotherapy treatment for colon cancer.
Team member(s)
Priyanka Parmar
Advisor(s)
Ansu Perekatt
Project name
Analysis of the fragmentation mechanisms of formic acid esters using GC-MS
Description
The project will focus on the analysis of mass spectra of several formic acid esters (formates). Especially phenylmethyl formic acid ester (benzyl formate), its mass spectrum includes a strong signal at m/z=90 and a notable signal at m/z=107. The hypothesis of the fragmentation of benzyl ester will be investigated in the research based on the previous confirmation on the presence of phenylcarbene radical cation, which contributes to the peak with m/z=90. As the same as the research done by Yaksha Jain, the benzyl formate substituted by deuterium will be used to predict the reaction mechanisms, and fragmentation behavior of benzyl formate, and will be generalized to apply to formic acid esters.
Team member(s)
Jia Chen
Advisor(s)
Athula Attygalle
Project name
Assessing Natural Killer Cell Cytotoxicity Against Multiple Myeloma Cells in the Presence of Interleukin-6
Description
Multiple myeloma (MM) is a blood cancer which affects plasma cells, a type of white blood cell. The proliferation of abnormal plasma cells in the bone marrow leads to a lack of production of healthy antibodies to recognize and fight infections. Malignant plasma cells in MM produce unusable antibodies that accumulate in the body. Therefore, MM can result in complications including fragility of the bones, anemia, kidney failure, higher susceptibility to infection, and various other effects to the body systems. While MM remains to be an incurable form of cancer, research into therapies such as the use of natural killer (NK) cells suggest their possibility for a treatment for multiple myeloma. NK cells are naturally produced lymphocytes which can recognize and attack malignant cells without prior activation or recognition, making their functions highly valuable to study. Additionally, cytokines produced by the body, such as interleukin-6 (IL6) are natural inflammatory response substances and are suspected to hinder cytotoxic activity for NK cells. For this reason, the interactions between NK cells and MM in the environment of IL6 and their production are of interest in understanding the optimal conditions which maximize NK cell cytotoxicity for its use as an effective multiple myeloma treatment.
Team member(s)
Malak Aziz
Advisor(s)
Woo Lee
Project name
Beyond Medicine: Direct Advertising to Customer-Patients
Description
Since its legalization in 1997, direct-to-consumer advertising of pharmaceutical drugs has been widely debated. Despite protests from pharmaceutical companies that it assisted patients with additional information regarding their care, a large portion of past research indicated potential harm to the relationship between patients and their care providers. There was a large gap in the information presented in direct advertisements and what a provider deemed necessary for patients to know. Additionally, pharmaceutical companies used manipulative techniques to boost sales and increase customer-patient interest in their products. To combat this and help inform customer-patients, we created a guide on how to navigate the pharmaceutical advertising world, complete with an explanation of the techniques used by Big Pharma. Information gathered through surveys and interviews, as well as investigation into past studies, was used to create this guide. By having this information at their fingertips, customer-patients could identify the deceptive techniques used by pharmaceutical companies. The guide aimed to educate customer-patients on information pertinent to their care from these advertisements, presenting a valuable resource for those seeking transparency in pharmaceutical marketing.
Team member(s)
Jordan Casey, Brooke Delo, Elizabeth Patterson, Gillian Reinhardt
Advisor(s)
Donald Lombardi, Patricia Anne Muisener
Project name
Biocidal Activity of Peroxide Radicals Generated from a Functionalized Nano-FeOOHSystem
Description
Bacterial infections can develop from medical implants and devices, posing a serious and life-threatening problem for patients. Approximately 32 million Americans have an implanted medical device, and they are at risk to develop a multitude of bacterial infections. Staphylococcus aureus is a gram-positive bacteria and one of the main causes of orthopedic-implant associated infections. Major infections including septic arthritis and osteomyelitis can arise, resulting in inflammatory destruction of joints and bone. To combat biofilm formation, metal oxide nanoparticles can be employed as a powerful anti-microbial surface modulator in combination with argon plasma due to the enhanced generation of peroxide radicals and the redox dynamic of dopamine and polydopamine. In this project, iron oxyhydroxide (FeOOH) nanoparticles were synthesized and tested on the growth of S. aureus. The metal nanoparticles were synthesized with a polydopamine (PDA) coating and an argon plasma reduction method. The results of this study revealed that the PDA-Fe coated fabrics treated with argon plasma exhibited a significant antibacterial activity against S. aureus. The importance of this research is to better understand the effects of metal nanoparticles as antimicrobial agents, to lower the rate of medical device-associated infections and prevent the increase of antibiotic resistance.
Team member(s)
Ameenah Aref, Shanel Calle Urgiles, Breanna Tolocka
Advisor(s)
Junfeng Liang, Jue Wang
Project name
Design and Synthesis of Metabolically Stable COVID-19 Oral Compounds with the Use of Machine Learning and Artificial Intelligence
Description
When COVID-19 hit the world, there was a search for a treatment for the virus in infected patients. Pfizer patented Nirmatrelvir in 2021, a protease inhibitor that stops viral replication in the body and can be taken as an oral treatment for COVID-19. Due to Nirmatrelvir being metabolized in the body too quickly, it is paired with Ritonavir, which slows the molecule's metabolism. The problem with this is that other drugs taken by the patient will also have a slowed metabolism which can alter their effectiveness. To find an analog to Nirmatrelvir that could inhibit the replication of COVID-19 and be metabolically stable as to be compatible with other essential medications, computer programs were used to screen designed molecules to predict their ability to dock to the protease. To have a metabolically stable COVID-19 inhibitor would mean that patients who contract COVID and may be at risk of the virus giving them health issues can take this drug orally without halting their other essential medications. During this research, a molecule was designed with blocked metabolic sites while keeping key interactions, selected based on its docking score, and the synthesis was performed in the lab.
Team member(s)
Alexis Pope, Krista Scartozzi, Mihika Shah
Advisor(s)
Sunil Paliwal
Project name
Development of Novel UBR Ligands — Using Computational Tools
Description
The ubiquitin proteosome system (UPS) plays a crucial role in the regulation of many cellular processes due to the degradation of misfolded or unneeded proteins, playing a role in various diseases and cancers. A crucial part of the UPS is E3 ligases which perform the catalytic transfer of ubiquitin to proteins for their degradation. Through this study, the UBR-box domain of the UBR family of E3 ligases was studied to computationally test and produce potent, small molecule ligands of the UBR5 E3 ligase. Based on a tetrapeptide previously found to potently bind to the UBR-box, arginine-isoleucine-phenylalanine-serine, novel compounds were produced to mimic its binding and were docked using Schrodinger Maestro software. Multiple rounds of compound modification, using bioisosteres and other moieties, and analysis of the docking scores and electrostatic interactions between the ligand and protein were performed. From the data collected, it was discovered that five novel compounds produced docking scores better than that of the tetrapeptide from literature. Through future research, including the synthesis and biological testing of these compounds, these novel ligands may be used for the treatment of diseases or other medicinal chemistry purposes, such as the production of novel proteolysis targeting chimeras(PROTACs).
Team members
Austin Ruth
Advisor(s)
Abhishek Sharma
Project name
Dye Degradations Through Catalytic Activity of β-FeOOH Nanoparticles
Description
Common sources of pollution stem from manufacturing, especially that of the clothing industry. The dye and other runoff often end up in water sources, where they can harm local ecosystems. In regions where manufacturing is common, such contamination can have a significant effect. To target the pollution, chemical catalysts are tested for their effectiveness in degradation. Dyes in labs can stand for manufacturing pollutants and are tested under a variety of conditions to discover how to safely mitigate the harmful effects of the pollutants. This research focused on the use of β-FeOOH nanoparticles. Ferric nanoparticles are used due to their non-toxicity, ease of creation, and efficacy as a catalyst in conjunction with hydrogen peroxide. When combined, they prove to be an effective method of degradation. Further testing of different environmental conditions, including heat, a common manufacturing byproduct, has proven to be effective in degrading the test dye methylene blue. Similar experiments have been completed utilizing various types of dyes and nanoparticles to approach the problem of dye pollution. Chemical catalysts pose as a safe and powerful tool in the solution to treating wastewater.
Team member(s)
Jennifer Przybysz
Advisor(s)
Junfeng Liang
Project name
Effects of SLC6A6 Attenuation on Ovarian Cancer Precursor Cells
Description
Non-ciliated fallopian tube epithelial (FNE) cells are precursor cells for the most frequently occurring type of ovarian cancer, high-grade serous ovarian cancer (HGSOC). Tumor protein p53 (TP53) is nearly ubiquitously mutated in ovarian cancers, and it is frequently associated with FNE cell transformation. How mutant p53 contributes to HGSOC development and progression is not well understood. Thus, we wanted to determine the consequences of mutant p53 expression in FNE cells. Previously conducted mass spectrometry of cell surface proteomics from our lab suggests that mutant p53 FNE induces expression of SLC6A6 taurine transporter, indicating the possibility that taurine transport mechanisms are involved in FNE-m-p53-dependent transformation. To investigate this possibility we used shRNA to attenuate SLC6A6 expression levels. Various phenotypical changes such as decreased proliferation and morphology changes were observed. Furthermore, we have demonstrated that lower SLC6A6 expression increases sensitivity to genotoxic stress evoked by cisplatin, a commonly used chemotherapy agent for HGSOC. With these findings, we plan to further validate these phenotypical changes in multiple ovarian cancer cell lines and move on to a mechanistic study.
Team member(s)
Teagan Polotaye
Advisor(s)
Marcin Iwanicki, Daniel Centeno
Project name
Environmental Impacts of Sunscreens Containing Zinc Oxide and Titanium Dioxide Nanoparticles on Marine Life
Description
Humanity today is facing an environmental crisis. One of the issues the world is facing is water pollution and the destruction of marine ecosystems. Every year this problem is exacerbated when millions of beach goers contaminate the oceans with sunscreen. Sunscreen usage is necessary for protecting against skin cancer but is causing immeasurable harm to marine life. Recent efforts have been made in an attempt to mitigate these issues. One such effort was the Hawaii Act 104, that as of 2021 banned sunscreen containing oxybenzone and octinoxate from sale in Hawaii. Instead of these organic UV filters, sunscreen manufactures have turned to inorganic UV filters like zinc oxide or titanium dioxide nanoparticles for sun protection.
Unfortunately, these UV filters have not proved to be the cure-all for marine life and new studies have been exploring the negative effects of inorganic UV filters. But which of these inorganic UV filters causes the most damage to marine ecosystems? This study compares data found in the literature from different experiments studying the effects of zinc oxide and titanium dioxide, to try to determine which sunscreen is the most detrimental to our environment. Determining the least harmful UV filter to use would assist in minimizing environmental damage allowing time for scientists to pursue nontoxic alternatives.
Team member(s)
Hannah Dicovitsky
Advisor(s)
Patricia Anne Muisener
Project name
Evaluation of Olipass Reagents Efficacy in Targeting SLC6A6 in Ovarian Cancer Cell Lines
Description
Non-ciliated fallopian tube epithelial (FNE) cells are precursor cells for the most common ovarian cancer, high-grade serous ovarian cancer (HGSOC). Solute carrier 6A6 (SLC6A6) mediates cellular taurine uptake. Our findings demonstrate that taurine confers cisplatin resistance in ovarian cancer cell lines and that SLC6A6 is expressed in cisplatin-resistant cells. This highlights SLC6A6 as a potential therapeutic target to enhance cisplatin sensitivity in ovarian cancer. Collaborating with OLIPASS Inc., we are developing strategies to target SLC6A6 in cell culture. Their Olipass Peptide Nucleic Acid (OPNA) technology modulates mRNA splicing to alter protein expression. Through western blot we have successfully knocked down SLC6A6 at a concentration of 1 uM OPNA in FNE cells. We are currently evaluating the efficacy of multiple SLC6A6-targeting OPNAs on several OC cell lines in targeting SLC6A6 downregulation.
Team member(s)
Paulina Georgoutsos
Advisor(s)
Marcin Iwanicki, Daniel Centeno
Project name
Investigating the Anti-Tumor Properties of Natural Killer Cells
Description
Multiple myeloma (MM) is a blood cancer that affects plasma cells, a type of white blood cell. The proliferation of abnormal plasma cells in the bone marrow leads to a lack of production of healthy antibodies to recognize and fight infections. While MM remains an incurable form of cancer, research into therapies such as the use of natural killer (NK) cells suggests their possibility for a treatment for multiple myeloma. NK cells are naturally produced lymphocytes that can recognize and attack malignant cells without prior activation, making their functions highly valuable to study. This project leverages the ability of different lines of NK cellsincluding iPSC-derived iNK cells, NK92 cells, and NK92MI cells to recognize and kill tumor cells. In addition to the multiple myeloma line, MM.1S, the NK cell's anti-tumor properties were studied against an ovarian cancer line, HEYA8 cells, and in a triple culture with ZT lung mesothelium cells, mimicking a tumors microenvironment. To assess NK cell cytotoxicity, this study assesses cultures of NK-92MI cells, iNK cells, ZT cells, HEYA8 cells, and MM.1s cells through imaging, ELISA, and Luciferase assays.
Team member(s)
Nicholas Housel
Advisor(s)
Woo Lee
Project name
Modeling Intracellular Concentrations of Free Amino Acids in Cultured Ovarian Cancer Precursor Cells
Description
Cancer cells accumulate free amino acids from the tumor microenvironment to support cell growth, proliferation, and survival. Profiling of the free amino acid content of ovarian cancer (OC) tumors derived from patient ascites revealed that taurine and proline, both osmolytes, were present at high concentrations (>100uM). Previous work in the Iwanicki Lab has uncovered that supplementation of taurine in culture media can restore intracellular taurine levels to those found within tumor samples. Accumulation of taurine has been shown to impede the proliferation of fallopian tube non-ciliated epithelial cells expressing mutant p53 (FNE-m-p53), a cell culture model representing OC precursors. Taurine’s antiproliferative effect was attributed to restoration of the tumor-suppressor p53 pathway and activation of the cell-cycle inhibitor CDKN1A (p21). Whether proline supplementation produces similar effects in FNE-m-p53 cells has not been explored.
The purpose of this investigation was to determine the effects of taurine and proline supplementation on cultured cells using a variety of assays, including flow cytometry, western blot, and live cell imaging. Our work has shown that proline, like taurine, activates p21 in cell lines expressing either mutant or wild-type p53. Interestingly, only taurine induced cisplatin resistance in FNE m-p53 cells. Furthermore, both amino acids induced changes in cell morphology, including modulation of cell size and adhesion through fibronectin production.
Team member(s)
Michael Greenberg
Advisor(s)
Marcin Iwanicki, Daniel Centeno
Project name
Mechanistic Investigation of Sustainable Heme Carbene N-H Insertions - Carbene Substituent Effect
Description
A combination of experimental research confirming the efficacy of iron porphyrin carbenes, or IPCs, as sustainable, alternative catalysts for N-H insertion reactions and computational research investigating the mechanisms of cyclopropanationreactions with IPC catalysts encourages the use of computational techniques to further investigate the mechanisms of heme carbene N-H insertion reactions. Empirical evidence from previous experimental research has determined that the structure of the ester substituent (R) of a particular asymmetric heme carbene N-H Insertion reaction which features an ester group on the carbene has significant effects on reaction yield and enantiomeric excess. However, more detailed insight into this mechanism is limited, so the primary intent of this research was to perform a computational mechanistic investigation of the key species of six reactions in this category with four different ester substituents (R = Et, Bn, Bn-OMe, and Bn-CF3) in order to compare their reaction energy barriers. The last two reactions were performed for both possible pathways (Ylide or HAT), each with the respective, energetically optimal axial ligand (L = His or Tyr) per pathway, which was previously determined by empirical results employing radical trappers and was also computationally investigated and verified.
Team member(s)
Catherine Minteer
Advisor(s)
Yong Zhang
Project name
Micro/Nano-Textured pHEMA Hydrogels for Optical Applications
Description
The problem that prompted this project was the limited efficiency of traditional optical devices for implantation and biomedical applications. The problem affects those that need better optical medical devices than those traditionally and widely available. This project introduces a groundbreaking solution through the development of pHEMA micro/nano-textured hydrogels. These hydrogels, featuring precise surface engineering at the micro and nano scales, can be more anti-microbial as well as much more biocompatible. PHEMA hydrogels trigger minimal immune responses and have cell-controlled degradation. The impact of this innovative solution allows for the use of polymeric materials with many unique qualities. These qualities include water absorption and retention, flexibility, biocompatibility, and responsiveness to environmental stimuli. Depending on the composition, hydrogels can be transparent, allowing for optical applications. This transparency is valuable in fields such as optics, where hydrogels can be used to minimize light scattering and enhance optical clarity. The adoption of pHEMA micro/nano-textured hydrogels allows for benefits for users in its applications.
Team member(s)
Alexandra Wong
Advisor(s)
Chang-Hwan Choi, Junfeng Liang
Project name
Microplastic Removal from Water
Description
Microplastics, particles less than 5 millimeters in length, are continually spreading across the environment. In addition to polluting the environment, microplastics cause many unintended consequences that will exist for centuries due to the material’slong degradation time. Marine life is affected by this growing issue especially sincemicroplastics can disrupt nutrient and carbon cycling. In addition, environmentalists, water treatment facilities, and the public, who rely on clean water sources, are also impacted by this pollution. Reducing human consumption of microplastics is critical as they can penetrate biological membranes, possibly impacting health. The problem at hand involves developing effective strategies for microplastic removal from water. To address this challenge, innovative solutions are being explored and researched, with a notable approach being the use of filters. In this study, a variety of filters was created and studied. One filter material was methylcellulose, which is a common material employed as a thickener and emulsifier in food and cosmetic products. To test the efficiency of the filters, stock solutions of known concentrations of various types of microplastics in addition to bottled water were filtered. The filters were then examined with different techniques which included infrared spectroscopy. In this presentation, we will present the results of our studies and compare the different filters. The impact of successful microplastic removal is far-reaching as it not only mitigates potential harm to aquatic ecosystems but also safeguards humanhealth. This research represents a promising step toward achieving cleaner water sources and fostering sustainable environmental practices.
Team member(s)
Kaelan Chaubal
Advisor(s)
Patricia Anne Muisener
Project name
Novel Mouse Models for Studying Kidney Cancer
Description
Kidney cancer ranks ninth among human cancers and over 14,000 patients succumb to the disease each year. There are a limited number of genetically engineered mouse models of kidney cancer. The majority of patient-derived xenograft (PDX)models do not mimic the in vivo environment, given that the xenografts are in immunocompromised mice. To develop a mouse model of kidney cancer, we simultaneously deregulated Wntand TGF-β signaling using conditionally mutant mice. It is known that Wnt signaling is associated with cell migration, proliferation, stem cell regeneration, renewal, and apoptosis, and it is also known that dysregulation of this pathway can lead to tumor growth. H&E staining and collagen staining was used to view the structure and compare the wild type with treated mutants, and we have begun performing IHC, in which we utilized the GFP biomarker to determine the presence of stem cells. We found expression of stem cell markers and phenotypic changes suggesting tumorigenic phenotype, and we are currently evaluating the markers associated with tumorigenesis in the kidney by immunohistochemistry.
Team member(s)
Julianna Bruzzese, Rohit Patel
Advisor(s)
Ansu Perekatt, Shima Nejati, Zahra Hashemi
Project name
Silver-Coated Fabric for Healthcare
Description
This study explores preparation and antibacterial performance of an antibacterial surface coated on fabric surfaces through PDA-silver-based coating. PDA-coated surfaces express many advantages such as good biocompatibility. The unique properties of dopamine, specifically its ability to self-polymerize, but the reaction is fast and uncontrolled. Glucose/borax plays a crucial role in the PDA coating via silver coating to control the reaction speed. By comparing the solution sample of GPDA (Glucose PDA) and BPDA (borax PDA) after 24 hours of incubation in different concentrations, we find out that glucose is a better copolymer that only affects the dopamine polymerization speed; while borax has concentration-breaking point, involved in a more complex mechanism which needs further study. This study focuses on GPDA for further modification by synthesizing silver coating and antibacterial applications. Antibacterial effects of PDA-silver coated fabric surface were then examined by placing the surface in bacterial solution and bacteria-containing agar plate. The result shows stronger antibacterial effectiveness of GPDA silver-coated fabric than silver ion does.
Team member(s)
Jiehao Zheng
Advisor(s)
Junfeng Liang, Runlin Li
Project name
The Beauty of Knowing Yourself: A Physiological Look into Dream Skin Care
Description
The success of skin care products on the current market is not only dependent on the scientific pathways of the product’s active ingredients’ effects on the skin. The product’s success is also contingent upon media marketing strategies and business tactics. Consumers typically purchase skin care products based on a trial-and-error process, due to a lack of understanding of the microbiome of their skin. This study combined modern research and literature reviews on the human skin microbiome and commercial applications to help users understand which products they should purchase based on the specific ingredients in those products. The demand for customized products is rapidly increasing in the skin care market. Therefore, this project proposed a feasible solution suggesting that a new design and personalized selling strategy should be implemented to help individuals make smarter purchasing decisions. The product idea set forth in this study references the skin microbiome and predicts buyer eligibility through skin microbiome tests. The overall purpose of this study was to stress the importance of becoming an “aware consumer.” With an understanding of the physiological relationship between skin care products and the skin microbiome, consumers can make more informed purchasing decisions in the absence of a licensed professional.
Team member(s)
Gianna Callegari, Aisha Hussain
Advisor(s)
Sarah Goodman
Project name
The Effects of Perfluorooctane Sulfonate (PFOS) on Transcriptional Reprogramming in Normal Fallopian Tube Epithelial Cells
Description
Perfluorooctane sulfonate (PFOS) is a man-made chemical once prized for its industrial applications due to its thermal stability and water-repellent properties. However, it is a persistent environmental contaminant that resists natural degradation and accumulates in human tissues. Given its ability to bioaccumulate, investigating PFOS's potential effects on gene regulation in epithelial cells, which are present in many organs, is crucial. To understand how PFOS exposure influences gene regulation, we employed a combination of human fallopian tube non-ciliated epithelial (FNE) cell cultures, high and low PFOS dose exposures, mass spectrometry to quantify cellular PFOS uptake, and transcriptomics to analyze gene expression changes. Our experiments yielded the following key findings: 1. Dose-Dependent Intracellular Accumulation: PFOS readily accumulated within the intracellular pool of molecules in FNE cells. Importantly, the level of accumulation was directly dependent on the strength of the exposure, with higher doses leading to greater intracellular PFOS concentrations. 2. Differential Gene Regulation: Exposure to varying PFOS concentrations resulted in the differential activation of gene transcription within the FNE cells. This suggests that the specific genes affected by PFOS depend on the level of exposure. These findings demonstrate that PFOS exposure can alter gene regulation in FNE cells, and the effect is dependent on the dose. We are currently elucidating the specific genes and pathways targeted by PFOS at different concentrations.
Team member(s)
Adam Leszczynski
Advisor(s)
Marcin Iwanicki, Tonja Pavlovic
Project name
The Role of Taurine in Age-Related Macular Degeneration (AMD)
Description
Taurine, a non-essential amino acid that is found throughout the body, has recently been linked to protective, anti-aging effects. Taurine is found in high concentrations in the Retinal Pigment Epithelium (RPE) cells, which serve as a barrier, supporting and maintaining the overlying photoreceptor cells. Age-related macular degeneration (AMD) is a disease characterized by RPE and photoreceptor cell degeneration and is the leading cause of blindness in individuals 55 and older. Mutations in the taurine transporter result in an early onset of AMD, suggesting taurine’s role in keeping RPE cells healthy and functional. When supplemented into in-vitro RPE cell culture, taurine was seen to have a positive effect on proliferation. Sodium iodate, an oxidant that is known to induce retinal degeneration through RPE cell death, was added to the in-vitro RPE cell culture in varying concentrations alongside taurine supplementation. Thus, we aim to investigate whether introducing taurine to the AMD-induced RPE cells may prevent further disease progression, and possibly reverse the harmful effects of sodium iodate. If successful, this experiment may offer a potential treatment candidate, taurine, for this debilitating disease.
Team member(s)
Maria Mihalatos
Advisor(s)
Marcin Iwanicki, Tonja Pavlovic
Project name
Unionization and Modern Healthcare & Industry: Fact versus Fiction
Description
This research aimed to address the following questions: (1) What differentiates unions from non-unions? (2) How do various perspectives on strikes influence our understanding of their impacts? (3) What are the actual impacts of strike action periods? (4) What are the ethical implications and justifications surrounding strikes? The primary focus was on determining the impact of strike actions in the healthcare and science-related industries. The findings revealed that strikes did not significantly affect patient mortality, but they did have a substantial impact on healthcare utilization, leading to significant declines in emergency room admissions, elective surgeries, and increased cancellations of outpatient appointments during strike periods. The complexity of strikes was acknowledged, due to their unique nature, and the call for necessary comprehensive longitudinal research following strikes was illuminated to gain a clearer comprehension of the true effects of such periods. Through the development of an educational piece, this research aimed to combat those who lack trust in a healthcare organization’s ability to uphold a high quality of care during strike periods.
Team member(s)
Michella Chiaramonte, Laura Masciola
Advisor(s)
Donald Lombardi, Patricia Muisener
Graduate Research Projects
Meet the Graduate Research Project Teams.
Project name
Addition of Superoxide Radical Anion to Halophenols Under Negative-ion Atmospheric Pressure Chemical Ionization Conditions
Description
The study focuses on the oxidative ionization of hydroxy group substituted aromatic compounds by superoxide radical anion (O2•-) under negative-ion superoxide conditions. The research found that the [M+ O2] •- adduct anion of iodophenol was present in considerable abundance, with a peak at m/z 252 in mass spectra acquired under negative-ion atmospheric pressure chemical ionization (APCI-) conditions. Further investigations confirmed that para, meta, and ortho isomers of iodophenol all undergo oxidative ionization and differ in relative adduct yield following the order of para-meta-ortho under identical conditions. Comparative studies across halophenols revealed that iodophenol, among the meta-substituted species, exhibited a remarkable capacity to form the adduct anion under optimal conditions for oxidative ionization. Other compounds showed similar potential in forming [M+ O2] •- adduct anions.
Team member(s)
Siyuan Li, Athulla Attygalle
Advisor(s)
Athulla Attygalle
Project name
An Approach of Anticancer Drug Delivery based on Controlled-Polydopamine Decoration
Description
Most anticancer drugs have very low solubility in water and lead to potential absorption problems. Orlistat, an antiobesity drug recently proved to have anticancer effect, which is extremely hydrophobic with very low efficacy of absorption. For more effectively delivering highly-hydrophobic drugs, we developed a simple approach to use tetraborate (borax)-controlled polydopamine and glucose-controlled polydopamine to bind with drugs and form a hydrophilic outer coating. Borax and glucose are both for controlling polymerization of polydopamine, which can generate more fine particles and intermediate 5,6-dihydroxyindole (DHI), providing a higher potential binding ability for drug and particles. In this research, orlistat and nile red, both very hydrophobic, were taken as two typical targets for determining the interaction between drugs and polydopamine particles. Particles were directly compared by visible dispersion change of drugs in solution and following characterized by dynamic light scattering and UV-vis spectroscopy. In addition of drugs to polydopamine particles, the anticancer cell viability test also showed a comparable result to free-drug ones. Taken those results together, our approach presents controlled polydopamine decoration is an ideal candidate for applications of drug delivery and cancer treatments.
Team member(s)
Runlin Li
Advisor(s)
Junfeng Liang
Project name
Assessing Natural Killer Cell Cytotoxicity Against Multiple Myeloma Cells in the Presence of Interleukin-6
Description
Multiple myeloma (MM) is a blood cancer which affects plasma cells, a type of white blood cell. The proliferation of abnormal plasma cells in the bone marrow leads to a lack of production of healthy antibodies to recognize and fight infections. Malignant plasma cells in MM produce unusable antibodies that accumulate in the body. Therefore, MM can result in complications including fragility of the bones, anemia, kidney failure, higher susceptibility to infection, and various other effects to the body systems. While MM remains to be an incurable form of cancer, research into therapies such as the use of natural killer (NK) cells suggest their possibility for a treatment for multiple myeloma. NK cells are naturally produced lymphocytes which can recognize and attack malignant cells without prior activation or recognition, making their functions highly valuable to study. Additionally, cytokines produced by the body, such as interleukin-6 (IL6) are natural inflammatory response substances and are suspected to hinder cytotoxic activity for NK cells. For this reason, the interactions between NK cells and MM in the environment of IL6 and their production are of interest in understanding the optimal conditions which maximize NK cell cytotoxicity for its use as an effective multiple myeloma treatment.
Team member(s)
Malak Aziz
Advisor(s)
Woo Lee
Project name
Determine Molecular Basis for the Attenuated Colitis Response Observed in Mice with Epithelia-Specific Loss of SMAD4
Description
Intestinal fibrosis is an understudied complication of IBD marked by the excessive accumulation of collagen and other ECM components within tissues. In the presence of persistent injury and/or inflammation, activated fibroblasts secrete ECM components and cytokines that modulate the immune response, thereby effecting the fibrotic response. The Transforming Growth Factor (TGF)-β1 is considered as a major mediator of fibrosis in the gut, impacting fibroblast proliferation. SMAD proteins act as the intracellular effectors of TGF-β1. Inflammation is a key driver of fibroblast activity and collagen deposition, which might lead to pathological wound healing. Non-pathological wound healing, hence requires proper collagen turnover. Differentiating between the accumulation of basement membrane collagen and interstitial matrix collagen is important in the context of fibrosis. The 40 K Da-weight DSS induces inflammation by ultimately disrupting the epithelial barrier function resulting in epithelial access to commensal bacteria, which are normally restricted to the outer mucus layer. To determine the cause of attenuated fibrotic response to DSS in the Smad4 knocked out (Smad4KO) colon, we assessed expression of collagens, proteins regulating epithelial integrity, repair response and responsible growth factors and pro-/anti-inflammatory immune cells in the Smad4KO colon versus the colon from normal mice after DSS treatment. We find a significant increase in the rate of epithelial migration in the Smad4KO mice following DSS treatment and differences in the infiltration of immune cells such as FOXP3 Treg cells as an anti-inflammatory cell in response to DSS. In Smad4KO mice, there is an enhanced wound healing response alongside an increase in the deposition of epithelial-specific collagen, even though the overall cumulative collagen deposition is lower compared to the wild type of colon. The research aims to investigate the impact of Smad4 loss on the fibrotic response to DSS treatment. The hypothesis is that Smad4 loss modifies the cellular and cytokine environment to attenuate fibrosis. This study’s findings will enhance our understanding of intestinal fibrosis in IBD and potentially lead to new therapeutic strategies against fibrosis.
Team member(s)
Zahra Hashemi
Advisor(s)
Ansu Perekatt
Project name
Development of Novel CDK 4/6 degraders
Description
Cyclin-dependent kinases 4 and 6 (CDK4/6) have recently emerged as major therapeutic targets for the treatment of breast cancer. CDK 4/6 inhibitors (CDK 4/6i) such as Palbociclib and Ribociclib were approved by US FDA for use in a combination therapy for estrogen receptor-positive, HER2-negative advanced breast cancer. However, patients often develop resistance to these drugs mainly due to kinase reactivation and decreased binding affinity of CDKi’s. To overcome these challenges, we have developed novel bifunctional PROTACs (Proteolysis Targeting Chimeras) for targeted degradation of CDK 4/6. Some of these molecules were found to be potent and selective degraders of CDK 6, thereby, inhibiting the phosphorylation of tumor suppressor retinoblastoma (Rb) protein.
Team member(s)
Aiswarya Mini, Qing Li, Sarat Chandarlapaty and Abhishek Sharma
Advisor(s)
Abhishek Sharma
Project name
Drug Resistance in Multiple Myeloma: The Role of Tumor Microenvironment and Stromal Interactions
Description
Despite advances in treatment, Multiple Myeloma (MM) remains incurable, partly due to the tumor microenvironment (TME), which significantly impacts drug efficacy. This study investigates the TME’s protective effects on MM cells against the drugs Bortezomib and Carfilzomib. MM cell lines were co-cultured with stromal cells—osteoblasts, endothelial cells, and fibroblast-like stromal cells—under conditions simulating the bone marrow environment, including fluid shear stress. Increased MM cell proliferation was observed, highlighting the role of stromal interactions and mechanical stress in protecting MM cells from these drugs. Preliminary RNA sequencing of MM (MM.1S) cells co-cultured with human fetal osteoblast (hFOB) cells identified significant gene expression changes in pathways associated with cytokine signaling and cell surface interactions, contributing to Cell Adhesion-Mediated Drug Resistance (CAM-DR). Validation of these findings is underway at both the RNA and protein levels. The research suggests that combining existing therapies with novel drugs designed to counteract the TME’s protective effects could enhance MM treatment efficacy.
Team member(s)
Panteha Behboodi
Advisor(s)
Woo Lee, Professor Benjamin Tycko, MD-Ph.D., Center for Discovery and Innovation (CDI)
Project name
ECM Regulation of Chemotherapy Resistance in Ovarian Cancer
Description
Ovarian cancer (OC) is a leading cause of death from gynecological malignancies. Although OC initially responds to chemotherapy, it often relapses and develops resistance, which complicates treatment efforts. Recent studies have linked the suppression of OC growth to cell protection against genotoxic drugs, yet the involvement of integrins and cell-matrix adhesion in these mechanisms is not fully understood. In our study using OC cell spheroid models, we demonstrated that the inhibition of cell proliferation by the CDK4/6 inhibitor Palbociclib results in cell protection from cisplatin. Bulk RNA sequencing of Palbociclib-treated OC cell line spheroids revealed the activation of ECM adhesion transcripts, including integrin β4. Analysis of The Cancer Genome Atlas (TCGA) data for ovarian cancer supported the link between increased ECM/integrin β4 mRNA expression and the presence of negative regulators of proliferation. In line with these findings, overexpression of integrin β4 and reconstitution of the ECM were enough to both suppress OC cell proliferation and enhance cell protection from cisplatin. Our results provide new evidence that cytostasis is associated with integrin-β4 and ECM-dependent cell protection from genotoxic stress in OC cell models.
Team member(s)
Sadaf Farsinejad
Advisor(s)
Marcin Iwanicki
Project name
Effects of Electrical Stimulation on Neural Cells for Tissue Regeneration Applications
Description
Peripheral nerve injuries (PNIs) represent a significant medical challenge, with severe cases affecting 44 individuals per 1 million annually and incurring an average cost of $48,000 per patient. Existing treatments, such as neurorrhaphy, nerve conduits, and autografting, offer limited success; even autografts, considered as the gold standard, have a success rate of only 40%. Electrical stimulation (ES) has emerged as a promising intervention to enhance nerve regeneration after surgery, yet its mechanistic regulation remains unclear. Understanding the underlying mechanism is critical for further developing innovative treatments of PNIs. In this study, voltage-gated calcium channels (VGCCs), which open in response to cell membrane potential changes, were hypothesized to be activated by ES, thus allowing for calcium influx and subsequent activation of intracellular signaling pathways and gene transcription within the nucleus.
To demonstrate the hypothesis, differentiated PC-12 cells were cultured on the tissue culture titer plate and a gap of 700um was created using the Incucyte 96-Well Woundmaker Tool. Through a customized device, ensuring sterility and functionality for repeated use in biological experiments without compromising the culture environment, 0 mV, 100mV and 150mV of direct current (DC) was applied to the cells daily and the gap closure was tracked via time-lapse imaging. In addition, a calcium probe was used to monitor the change of intracellular calcium concentrations while undergoing ES. Neurite branch lengths within each gap were evaluated using skeletonized images with FIJI/ImageJ software. Results of the gap study after 9 days show a median branch length of 29 +/- 7, 50 +/- 18, and 68 +/- 21 um for 0mV, 100mV, and 150mV ES groups, respectively. Furthermore, there was a trend in the change in intracellular calcium concentration between 0mV and 100mV ES groups. Notably, increased intracellular calcium levels were constantly observed in the ES groups, such that 100mV ES group exhibited the [Ca2+]i value of 24 +/- 9 while the 0mV group had a value of 8 +/- 6. These observations partially support the hypothesis that ES-induced electric potential changes of cell membrane trigger VGCCs to facilitate calcium entry and activate downstream regenerative processes. Calcium influx through VGCCs can influence the growth cone dynamics and guide neurite outgrowth, which is a critical step in the regeneration of damaged nerves (1). In summary, our results indicate that stimulation of PC-12 cells with ES led to elevated intracellular calcium concentrations while promoting the extension of neurites from the cell body. Initial investigations suggest that ES might mediate neuronal cells by altering membrane potential, which in turn triggers the opening of voltage-gated calcium channels, facilitating calcium entry into the cell. There is a clear trend on the occurrence of calcium influx when in the presence of DC stimulation. Clearly, identifying the underlying intracellular mechanism in response to ES would be essential, offering the opportunity to design novel therapeutics to promote peripheral nerve regeneration.
References
1. Kamijo, S. et al. J Neurosci 38, 5551–5566 (2018).
Team member(s)
Felicia Spadavecchia
Advisor(s)
Hongjun Wang
Project name
Epigenetic Reprogramming and Its Contribution to Multiple Myeloma Cell Plasticity and Drug Resistance
Description
The clinical management of Multiple Myeloma (MM) remains a significant challenge due to the development of drug resistance, which is influenced by the tumor microenvironment (TME). This resistance impedes the efficacy of standard therapies, calling for innovative approaches to treatment. This study investigates the potential of epigenetic modification, specifically DNA hypomethylation using the drug Decitabine (DAC), to enhance the treatment efficacy of Bortezomib in MM.
Our experiment involved culturing three MM cell lines—MM1S, NCI-H929, and U266—in both monoculture and in co-culture with human fetal osteoblast cells (hFOB) to simulate the TME. The MM cells were pretreated with 1 µM Decitabine for 72 hours to induce epigenetic reprogramming before exposure to Bortezomib. Preliminary results showed that the TME notably influenced the drug resistance in MM cells; MM1S and NCI-H929 demonstrated increased survival and proliferation when co-cultured with hFOB, an effect not observed with U266 cells. Furthermore, U266 cells exhibited a heightened sensitivity to Decitabine compared to MM1S and NCI-H929, likely due to genetic differences associated with the p53 gene.
Our data indicate that the utilization of DNA hypomethylation through Decitabine can improve the sensitivity of MM cells to Bortezomib. The combination of Decitabine and Bortezomib was more effective in reducing cell viability across all cell lines and culture conditions compared to Bortezomib alone. This suggests that targeting epigenetic mechanisms may offer a promising strategy to counteract drug resistance in MM. Future work will include comprehensive DNA methylation profiling and RNA sequencing to delineate the molecular mechanisms behind the observed effects, alongside RT-qPCR and Western blot analyses to confirm changes at the transcriptional and protein levels.
This approach represents a potential shift in treating MM by integrating epigenetic therapy to overcome resistance within the TME, thus improving the outcomes for patients undergoing chemotherapy.
Team member(s)
Sam Talaei
Advisor(s)
Woo Lee
Project name
Functionalized Gold Nanoparticles for Facile Pattern-Controlled Surface Coatings
Description
Gold nanoparticles (AuNPs) have been widely investigated as surface modifiers; nevertheless, most methods still require the pretreatment of surfaces and several steps to control coating efficiency and patterns for improved functionality. We developed functionalized AuNPs through borate-protected dopamine (B-AuNPs). The simple activation of B-AuNPs with a strong acid to remove the protected borate groups produces adhesive dopamine AuNPs (D-AuNPs). D-AuNP-coated surfaces with varied but controlled features and properties such as coating density and surface pattern were achieved using D-AuNPs with a precisely controlled dopamine density and coating conditions. Such adhesive and easily manipulated AuNPs provide a facile and time-saving technology to achieve sophisticated surface coatings using AuNPs.
Team member(s)
Jue Wang, Junfeng Liang
Advisor(s)
Junfeng Liang
Project name
Immune Dynamics During Intestinal Tumorigenesis
Description
Colorectal cancer ranks third in diagnosis frequency. Immune changes within the gastrointestinal tract as the largest immune organ impact tumor development and progression. Tumor cells evade immune surveillance by reprogramming immune cells to create a suppressive tumor microenvironment (TME), facilitating cancer progression. Among immune cells, myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs) and tumor-associated macrophages (M2) play a key role in cancer progression and immune suppression in the TME. Hence, the reciprocal signaling between the immune and tumorigenic cells is presumed to promote cancer progression.
Material and Method: To study the immune dynamics during tumor progression, we used an inducible mutant mouse model that mimics concomitant deregulation of Wnt and TGFβ signaling. Following Tamoxifen injection, mutant mice develop tumorigenic hyperplasia in the intestinal epithelium within three weeks. Immunostaining for immune markers associated with immunosuppression and pro-inflammatory responses assesses immune changes at this time point.
Results: The immune population associated with immunosuppression (MDSCs, Treg and M2 macrophages) increased while the pro-inflammatory population (iNOS-positive cells) decreased in the mutant intestine during tumorigenesis.
Conclusion: Tumorigenesis is accompanied by changes in the immune population that promote an immunosuppressive milieu, which might facilitate cancer progression.
Team member(s)
Shima Nejati
Advisor(s)
Ansu Perekatt
Project name
Intriguing Cooling Effects Enacted by Solvent Vapors on Intramolecular Proton Hopping in Gas-Phase Ion Ensembles
Description
In peptide mass spectrometry, the “Mobile Proton Model” describes how protons initially attached to more basic sites of a peptide can translocate to the backbone after ion activation. Similarly, in aromatic amines like aniline or amino naphthalene, a “ring walk” occurs via intramolecular proton transfer after ion activation. This study investigates the influence of solvent vapor presence in the ion source environment on intramolecular proton hopping. It was found that solvents significantly affect proton hopping, with the ion-mobility separated protomers of aniline and amino naphthalene, generated under high in-source activation, being dominated by the ring-protomer ©. Introducing vapors like methanol, acetonitrile, acetone, dichloromethane, ethyl acetate, tetrahydrofuran, or n-hexane into the ion source changed the mobilogram profile to be dominated by the amino group (N-protomer). The study revealed that hexane’s efficiency is similar to other solvents, suggesting that intermolecular associations between ions and solvent molecules are not the primary interactions mediating the observed changes. Instead, in the absence of solvent vapors, high-energy ions anneal towards thermodynamically favorable protomers © via intramolecular proton-hopping without external assistance. In contrast, the presence of vapor molecules hinders ions from overcoming transition energy barriers due to collisional cooling effects, retarding the intramolecular transformation. This discovery could lead to new methods for studying intramolecular proton hopping in peptides during mass spectrometric analysis.
Team member(s)
Meenu Kumar
Advisor(s)
Athula Attygalle
Project name
Investigating the Immunoregulatory Effects of Endothelial Cells in the Natural Killer Cell Cytotoxicity in Multiple Myeloma-Associated Tumor Microenvironment
Description
Natural Killer (NK) cells exhibit potent cytotoxicity against Multiple Myeloma (MM). However, leveraging NK cells as an adoptive cell transfer therapy for MM patients is challenged by the immunosuppressive MM tumor microenvironment (TME). Understanding the molecular interplay between MM cells and non-hematopoietic cells within the bone marrow is crucial for enhancing NK cell-mediated anti-myeloma activity.
We evaluated the effect of the presence of stromal cells (fibroblasts, osteoblasts, endothelial cells, and primary adipocytes) in co and triple-cultures with MM and NK cells. We found that all four types of stromal cells enhanced the growth of MM cells in vitro, but only some cell types conferred protection from NK cell-mediated cytotoxicity in triple culture experiments. The strongest NK cell resistance was observed in the presence of endothelial cells (human umbilical vein endothelial cells (HUVEC)). Analyzing NK cell activation in NK cell monocultures compared with co and triple culture conditions (NK cells +/- MM cells +/- HUVEC) revealed that both MM cells and HUVEC triggered NK cell activation, and HUVEC did not suppress NK cell activation. In a preliminary RNA-sequencing experiment, we found that co-culture of MM cells (MM.1S cell line) with HUVEC had a profound effect on the transcriptional profile of MM.1S cells with the following top 10 list of upregulated genes: ITGA5 (CD49a), SERPINE1, DKK3, CCN1, PRSS23, IL1RL1, COL5A1, SOCS3, THBS1, and HHIP, suggesting that HUVEC induced pro-survival genes in MM cells thus promoting resistance to NK cell cytolytic activity.
Collectively, our findings can pave the way for innovative strategies to enhance the efficacy of NK cell therapy. Future directions of this project will include validation of our RNA sequencing data, identification of pathways associated with immunotherapy resistance in MM, and the exploration of novel combinatorial approaches (combining NK cell therapy with small molecules or antibodies) to overcome TME-induced immunosuppression in MM.
Team member(s)
Shabnam Samimi
Advisor(s)
Woo Lee
Professor Johannes Zakrzewski, MD, Center for Discovery and Innovation, Hackensack Meridian Health, NJ.
Project name
Mechanistic Investigation of HNO Formation from Hydroxyurea Catalyzed by Horseradish Peroxidase
Description
Hydroxyurea is a well-established small-molecule drug used for the treatment of many myeloproliferative conditions, cancer, acquired immunodeficiency syndrome, and sickle cell anemia. It has been recently proposed that nitroxyl (HNO), also known as azanone, generated from hydroxyurea is the active species responsible for the treatment of sickle cell disease; however, the mechanism of action of hydroxyurea remains unknown. In this communication, we employed density functional theory (DFT) calculations to provide the first detailed mechanistic investigation of HNO generation from hydroxyurea catalyzed by hydrogen peroxide treated horseradish peroxidase (HRP). Our results reveal two subsequent hydrogen atom transfers (HATs) from hydroxyurea to HRP Compound I as the most favorable mechanism for HNO generation. Moreover, a large active site model was studied to determine the most favorable substrate conformation throughout the reaction. We demonstrate that the coupled effect of His42 side chain rotation and hydroxyurea’s interaction with the nearby W2240 water molecule significantly improves substrate’s reactivity. Overall, this study reveals, for the first time, the reaction mechanism of HNO generation from hydroxyurea, providing critical electronic, geometric, and energetic data not known before. This comprehensive theoretical investigation shall facilitate the future development of HNO donor drugs for broad biomedical applications.
Team member(s)
Dariya Baizhigitova, Erika McCarthy, Jia-Min Chu
Advisor(s)
Yong Zhang
Project name
Modeling of Intracellular Taurine Levels Associated with Ovarian Cancer Reveals Activation of p53, ERK, mTOR and DNA Damage Sensing-Dependent Cell Protection
Description
Taurine is known to protect various tissues from degeneration associated with the cancer therapeutic agent cisplatin. The protection mechanism of human ovarian cancer (OC) cells from DNA damage caused by cisplatin through taurine has not been previously explored. Ascites cells from chemotherapy-naive ovarian cancer patients were found to contain significantly higher levels of intracellular taurine compared to cell culture models of ovarian carcinoma. Elevating intracellular taurine to levels associated with ascites cells suppressed growth in various OC cell models, including chemotherapy-naive fallopian tube epithelial cells expressing mutant p53 (FNE-m-p53), multiple OC cell lines, and OC patient-derived organoids. This growth inhibition was linked to reduced glycolysis, decreased DNA damage, and enhanced cell protection from cisplatin. A combination of RNA sequencing, reverse phase protein arrays, live-cell microscopy, flow cytometry, and biochemical validation experiments indicated that taurine induces the binding of mutant- or wild-type p53 to DNA, activating p53 effectors that negatively regulate the cell cycle (p21) and glycolysis (TIGAR). Notably, taurine’s suppression of cell proliferation was associated with the activation of mitogenic signals such as ERK, mTOR, and an increase in mRNA expression levels of major DNA damage-sensing molecules like DNAPK, ATM, and ATR. While inhibiting ERK or attenuating p53 with shRNA did not affect taurine’s protective ability against cisplatin, inhibiting mTOR with torin2 (which also targets DNAPK and ATM/ATR) did impede taurine’s protective effect. The studies suggest that an increase in intracellular taurine could suppress cell growth, alter metabolism, and activate cell protective mechanisms involving mTOR and DNA damage sensing.
Team member(s)
Daniel Centeno
Advisor(s)
Marcin Iwanicki
Project name
Molecular Characterization of Dedifferentiation-Induced Oncogenic Stemness in the Intestinal Epithelium: A Smad4KO:β-CateninGOF Conditional Mutant Mouse Model
Description
The molecular characteristics that distinguish tissue-specific stem cells from cancer stemness remain underexplored. Our investigation focuses on the molecular events associated with dedifferentiation-induced oncogenic stemness within the intestinal epithelium, utilizing a Smad4 knockout:β-catenin gain-of-function (Smad4KO:β-cateninGOF) conditional mutant mouse model. The intestinal epithelium is spatially organized, delineating the proliferative crypt compartment from the differentiated villus compartment. The crypt compartment houses the stem cells which have been shown to be the cell-of-origin of colon cancer. However, cancer has also been shown to arise from the differentiated cells of the villus compartment. Thus, there are two models for the origin of colon cancer: the top-down model, wherein tumors arise from differentiated cells, and the bottom-up model, wherein tumorigenesis occurs from stem cells. The mouse model employed here mimics the top-down model of tumorigenesis and allows for the molecular characterization of cellular changes during the transition from differentiated cells to oncogenic stem cells. Upon induction of this mutation, we observe cell fate reversal to oncogenic stemness within the differentiated compartment. This cellular event is characterized by the aberrant activation of Notch signaling, a pathway normally restricted to the crypt compartment in healthy epithelium. Furthermore, single-cell RNA sequencing (scRNA-seq) analyses uncover a dual trajectory during this dedifferentiation towards stemness characterized by the emergence of two distinct populations: oncogenic stem cells and non-oncogenic stem cells. Sequencing analysis also reveals aberrant Notch activity and oncogenic markers during the dedifferentiation process. These findings suggest that anomalous Notch signaling in the differentiated compartment may be a driving force underlying the acquisition of oncogenic stemness in the differentiated compartment. Ultimately, this study provides valuable insights into the molecular mechanisms underlying dedifferentiation-induced oncogenic stemness in the intestinal epithelium. The implications of epigenetic changes and transcriptomic alterations in this process provide valuable avenues for further research into colorectal cancer pathogenesis and therapeutic intervention.
Team member(s)
Kylee Wrath, Amartya Singh, Thomson Hui, Zahra Hashemi, Steven Zukowski, Crystal Lim, Hossein Khiabanian, and Ansu Perekatt
Advisor(s)
Ansu Perekatt
Project name
Vinyl cyclopropane diborons: Versatile reagents to access homoallylic α,α-diboryl radicals
Description
α-Boryl radicals are an emerging and highly useful class of reaction intermediates in organic synthesis. These radicals are stabilized via empty p-orbitals on adjacent boron atoms. However, the synthetic application of these α-boryl radicals are underexplored. We have developed a vinyl cyclopropane diboron-based strategy to generate rare homoallylic α,α- diboryl radicals. These radicals smoothly undergo cis diastereoselective [3+2] cycloaddition reactions with various olefin partners. The mild reaction conditions also allowed a one-pot vinyl cyclopropane (VCP) ring-opening, cycloaddition-oxidation sequence to afford disubstituted cyclopentanones with good to excellent yields. Cyclopentane and cyclopropane motifs are ubiquitous in bioactive natural product and various FDA approved drugs. Control experiments and DFT mechanistic study confirmed a radical mediated reaction pathway. To our knowledge these are the first examples of the use of geminal diboryl group as an activator of VCP ring opening and cycloaddition reaction of α-boryl radicals.
Team member(s)
Het Vyas
Advisor(s)
Abhishek Sharma
Presenter
Jun Liu, Ph.D. Student/Candidate
Project name
Using Whole-body Linear Momentum Control Strategies to Understand Two-Foot Running Jumps with a Basketball vs. Without
Research cluster
Biomechanics and Rehabilitation Engineering
Department
Biomedical Engineering
Advisor(s)
Antonia Zaferiou