Louisiana Research Day Program Book 2025
Animated publication
2025 Via Research Recognition Day
1
2025 Research Recognition Day
ON THE COVER: (left to right) Grace Lee, OMS-II; Berony Geneste, OMS-II
Agenda February 28, 2025
Erin Vasasquez OMS-II
7:30–8:30 a.m.
Registration, second floor Coffee service and refreshments, second floor
8:30–8:40 a.m.
Opening Remarks Lecture Hall 2, second floor
Welcome
8:40–9:00 a.m.
Plenary Lecture: “The State of Research at VCOM” P. Gunnar Brolinson, DO, FAOASM, FAAFP, FACOFP Lecture Hall 2, second floor
Delaney Yu OMS-IV
Welcome to the VCOM Via Research Recognition Day on the VCOM Louisiana Campus. Each year, the Via Research Recognition Day is a significant event for VCOM that supports the mission of the College to provide medical education and research that prepares globally minded, community-focused physicians and improves the health of those most in need. The Via Research Recognition Day offers a forum for health professionals and scientists in academic institutions, teaching hospitals and practice sites to present and benefit from new research innovations and programs intended to improve the health of all humans. By attending the sessions with guest speakers, participants have the opportunity to learn cutting edge information in the physiological bases of osteopathic manipulative therapy efficacy, new trends in physician-based research networks, and how to develop innovative research projects with high impact for human health. Poster sessions allow participants to learn about the biomedical, clinical and education-simulation research activities at VCOM-Louisiana and its partner institutions. This year, the Biomedical Research category at the VCOM-Louisiana Research Day event has been expanded to include two sections: Section 1 will feature presenters primarily from VCOM, while Section 2 will showcase contributions from our partner institutions. This change reflects rapidly growing interest and participation in this event.
9:00–10:00 a.m.
Keynote Lecture: “Host Metabolic Status as a Key Determinant of Viral Disease Progression and Transmission” James Weger-Lucarelli, PhD Virginia Tech Lecture Hall 2, second floor
10:00–11:30 a.m.
Brunch, appetizer buffet First floor hallway between rooms 107 and 132
10:15 a.m.–12:00 p.m.
Poster Competition Rooms-107, 129-132, first floor
1:00–1:30 p.m.
Award Ceremony and Final Remarks Lecture Hall 2, second floor
2
3
2025 Research Recognition Day
RESEARCH
Contents
Speakers
Program Moderators.................................................................................................................................................................6
Plenary Speaker.........................................................................................................................................................................8
Keynote Speaker........................................................................................................................................................................9
Abstracts
Biomedical Research: Section 1.............................................................................................................................................10
Biomedical Research: Section 2.............................................................................................................................................33
Clinical Research.....................................................................................................................................................................42
Anatomy and Epidemiology....................................................................................................................................................51
Case Studies: Section 1..........................................................................................................................................................71
changes the world.
Case Studies: Section 2........................................................................................................................................................115
4
5
2025 Research Recognition Day
Program Moderator
Program Moderator
Supporters
Mark A. Sanders, DO, JD, MPH, LLM, MS, FACOFP, FACLM Dean, Louisiana Campus VCOM
Pawel Michalak, PhD Associate Dean for Biomedical Affairs & Research, Louisiana Campus VCOM
Mark A. Sanders, DO, JD, MPH, LLM, MS, FACOFP, FACLM, is the dean for VCOM Louisiana. Before becoming dean, Sanders held the position of associate dean for curriculum, assessment and medical education for VCOM-Louisiana, a role he had previously held at VCOM's Auburn campus. He continues to teach genetics, geriatrics, and ethics at VCOM. His research interests include topics in geriatrics, elder law, transitional care, chronic disease, and molecular genetics. He specializes in family medicine, geriatric medicine, legal medicine, and public health. He specifically caters to homebound elderly individuals and other adults with disabilities. Sanders graduated from Texas Christian University with a degree in biology/ chemistry. He then obtained his medical degree from UNTHSC-TCOM and his law degree from Texas A&M College of Law (formerly Texas Wesleyan University). He then received a Master of Public Health degree from UNTHSC-SPH and a master's
in elder law from Stetson University College of Law. Most recently, he obtained a graduate degree in clinical molecular genetics from Northern Michigan University. Dr. Sanders is a fellow in the American College of Osteopathic Family Practice and in the American College of Legal Medicine. In addition, he serves on the textbook editorial committee for LEGAL MEDICINE 2023, written by the American College of Legal Medicine. He authored a chapter in the same textbook on the use of artificial intelligence in genetics. Dr. Sanders is married to Anada Gunn Sanders, JD, MPH, and they have three children: Sofia, Liam, and Emma Grace.
Pawel Michalak, PhD, serves as the associate dean for biomedical affairs and research for the VCOM-Louisiana campus. He is also an adjunct faculty member at the Virginia Maryland College of Veterinary Medicine and the University of Haifa in Israel. Michalak served as a professor at the VCOM Virginia campus, with a focus on comparative genomics, bioinformatics, and One Health, operating as head of the Integrative Genome Analysis Lab (IGAL) at the Center for One Health Research. His team has employed an integrative approach with a variety of molecular, computational, and modeling techniques, including NextGen sequencing, a high-throughput DNA sequencing technology, of entire genomes, epigenomes, and transcriptomes to establish a rigorous understanding of how complexity elaborates from genomes through gene regulatory networks, and how it evolves over generations in response to environmental stress and intragenomic challenges.
Michalak has worked at nine universities in four countries across three continents. He has published his research in one book, with a second currently in preparation. Additionally, Michalak has been credited as a contributor in multiple books, as well as approximately 70 peer-reviewed papers in scientific journals, including “Cell,” “Science,” “Nature,” “Proceedings of the National Academy of Sciences USA” and “Proceedings of the Royal Society.” His research has been cited nearly 2000 times and popularized by various public media outlets.
Dixie Tooke-Rawlins, DO, FACOFP President VCOM
John Rocovich, Jr, JD, LLM Chairman VCOM
6
7
2025 Research Recognition Day
Plenary Speaker
Keynote Speaker
P. Gunnar Brolinson, DO, FAOASM, FAAFP, FACOFP Vice Provost for Research VCOM
James Weger-Lucarelli Research Assistant Professor Virginia Tech
James Weger-Lucarelli earned BS and PhD degrees from the University of Wisconsin Madison. He then performed postdoctoral studies at Colorado State University and the Pasteur Institute in Paris, France. His lab broadly studies virus-host interactions and virus evolution, with the goal of using this knowledge to design therapeutics, antivirals, and vaccines to prevent or treat disease. His lab has identified a causal link between metabolic disease and more severe disease outcomes upon viral infection. More recently, Weger-Lucarelli’s lab has begun applying knowledge of virus-host interactions and evolution to engineer viruses to kill cancer cells or reprogram the inflammatory environment to eradicate cancer.
P. Gunnar Brolinson, DO, FAOASM, FAAFP, FACOFP, is vice provost for research, professor of family and sports medicine at the VCOM and team physician for Virginia Polytechnic Institute and State University. He is an adjunct professor in the Department of Mechanical Engineering at Virginia Tech. He is the fellowship director emeritus of the Primary Care Sports Medicine Fellowship at VCOM and Virginia Tech. He is also a volunteer physician for the United States Olympic Committee and a team physician for the United States ski team and was head team physician for the freestyle ski team at the 2006 Winter Olympic Games in Torino, Italy. He was also named to the medical staff for the 2010 Winter Olympic Games in Vancouver, British Columbia, and was medical director of the USOC performance services center. He obtained his undergraduate training from the University of Missouri at Columbia earning a degree in biology. A 1983 graduate of the Kirksville
College of Osteopathic Medicine, Dr. Brolinson is board certified in family practice and holds a subspecialty certification in sports medicine. Prior to coming to Virginia, he was the co director of the Primary Care Sports Medicine Fellowship training program at The Toledo Hospital in Toledo, Ohio, and team physician for University of Toledo. He has extensive experience in undergraduate and post graduate medical education. Dr. Brolinson has served on the boards of the American Osteopathic Academy of Sports Medicine, the American Medical Society for Sports Medicine and the Midwest Chapter of the American College of Sports Medicine. He is a past president of the American Osteopathic Academy of Sports Medicine. Dr. Brolinson is a fellow of the American Osteopathic Academy of Sports Medicine, the American Academy of Family Practice and the American College of Osteopathic Family Practice. In 1997, he was named outstanding young physician in Ohio by the Ohio State Medical Association.
He is a former associate editor for the Clinical Journal of Sports Medicine and a former member of the editorial board of the “Physician and Sports Medicine.” He is former section editor for Competitive Sports and Pain Management in the journal “Current Sports Medicine Reports.” Dr. Brolinson is a frequent speaker at national sports medicine meetings and often teaches didactic laboratory sessions on the use of osteopathic manipulative therapy for athletic injuries. Dr. Brolinson is a contributing author in the latest edition of “Foundations for Osteopathic Medicine” and he has published several scholarly articles and book chapters in the area of sport and exercise medicine. His research interests have included exercise and immune function, exercise and bone mineral density, mild traumatic brain injury in sports, impact biomechanics, human factors in auto safety, sports performance and manipulation and other health and disease prevention related topics.
8
9
2025 Research Recognition Day
Biomedical Research: Section 1
Janika Ernst, BS ,OMS-IV 1 ; Arooj Arif BS, MS, OMS-II 1 ; Jourdan Blair, BS, OMS-III 1 ; Emily Gregory BA, MA ,OMS-II 1 ; Shailie Shah, BS, OMSII 1 ; Lin Kang, PhD 1 ; Robin Varghese, PhD 2 ; Melissa EH Lipsmeyer, MS, PhD 1 1 VCOM-Louisiana; 2 VCOM-Virginia 1 INDOLE-3-PROPIONIC ACID, A METABOLITE OF THE GUT-MICROBIOME, ALTERS NUMEROUS BIOLOGICAL PROCESSES OF PLACENTAL CELLS IN AN IN VITRO MODEL OF OBESITY
Context: Obesity during pregnancy is associated with detrimental changes in placental function that lead to the development of complications such as gestational diabetes and preeclampsia as well as altered metabolism in the offspring. With the incidence of obesity reaching epidemic levels world-wide, it is imperative to find mechanisms to mitigate these risks during pregnancy. The gut-microbiome is a diverse system that regulates numerous body functions through the secretion of specific metabolites. Gut dysbiosis has recently emerged as one mechanism that contributes to placental dysfunction and clear associations between obesity and gut dysbiosis are evident. Indole-3-propionic acid (IPA) is a gut microbiome derived metabolite that has anti-inflammatory, anti-oxidative stress and glucose regulatory functions, all of which are dysregulated in obesity. Previous studies have found altered levels of IPA in both serum and placentas of women with obesity. Additionally, it was found that serum IPA levels are correlated with lower levels of adiposity, circulating insulin and insulin resistance. However, it remains unknown what the physiological functions of IPA are in the placenta and if it could have potential protective effects against perturbations caused by obesity.
Objective/Hypothesis: The objective of this study is to determine what physiological role IPA plays in the placenta and if IPA can protect against obesity-induced alterations in cell function. We hypothesize that physiological levels of IPA will reduce inflammatory markers, alleviate oxidative stress and modulate glucose uptake through altered insulin signaling in placental cells based on previously documented functions of IPA in other tissue types. Methods: We developed an in vitro model of obesity where two human placental cell lines, BeWo (choriocarcinoma) and HTR8 (normal immortalized trophoblasts) were cultured in media conditioned by mature human adipocytes (ACM) to mimic an obesogenic environment. RNA sequencing (RNAseq) analysis was used to determine acute gene expression changes due to a 24 hour treatment with either a control (DMSO), IPA (1uM, physiological levels), ACM, or ACM+IPA to evaluate if IPA can reverse transcriptional changes induced by ACM. Results: Initial RNAseq analysis revealed ACM induced changes in genes related HIPPO signaling, and other metabolic pathways in
BeWo cells while changes in folate biosynthesis, DNA replication and cell cycle were altered with ACM+IPA treatment. In HTR8 cells, ACM treatment induced changes in inflammatory and extracellular-matrix-related pathways while ACM+IPA treatment altered focal adhesion, extracellular-matrix interactions and regulation of actin cytoskeleton function. Interestingly, the PI3K-AKT pathway was a common target in both cell lines with changes dependent upon the presence or absence of IPA. Conclusion: Our RNAseq analysis offers initial insight into the molecular mechanisms of IPA in placental cells that include alterations in extra cellular matrix dynamics and folate metabolism. These are novel functions of IPA not identified in other cell types. Collectively, this study highlights the importance of a healthy gut microbiome and potentially serves as a window of opportunity to mitigate the onset or severity of disorders such as gestational diabetes by enhancing the gut microbiome prior to pregnancy.
Biomedical Research: Section 1
10
11
2025 Research Recognition Day
Biomedical Research: Section 1
Biomedical Research: Section 1
A. Madi Grote, BS, OMS-III; J. Annelise Hayward, BS, OMS-III; Riva Kelly, BS, OMS-III; Caroline Naquin, BS, OMS-II; K Adam Morrow, PhD; Melissa EH Lipsmeyer, MS, PhD VCOM-Louisiana 2 INDOLE-3-PROPIONINC ACID MODULATES INSULIN SIGNALING IN ENDOMETRIOSIS CELLS IN VITRO
Context: Pseudomonas aeruginosa is an organism known to cause nosocomial pneumonia that can progress to sepsis and acute lung injury, especially in immunocompromised patients (Balzcon et. al. 2024, Stevens et. al. 2014). Following recovery from this infection, patients have elevated death rates and manifest various morbidities such as cognitive deficits (Balzcon et. al. 2023). Through a type 3 secretion system (T3SS) P. aeruginosa introduces exoenzymes into host cells, including exoenzyme Y (ExoY) during infection. ExoY-dependent production of cAMP activates protein kinase A (PKA) causing the hyperphosphorylation of endothelial tau, a multifunctional microtubule-associated protein (MAP) found in neuronal and endothelial cells. The phosphorylation of tau consequently impairs microtubule assembly (Baas and Qiang 2019, Morrow et. al. 2015). This cytotoxic tau can enter circulation making it a target of study for the diffuse effects seen following a nosocomial pneumonia infection (Balzcon et. al. 2023). Furthermore, tau has other properties beyond acting as a simple stabilizer of microtubules. Tau has been indicated in Alzheimer’s disease along with amyloid beta (A β ), a multifunctional protein Alexis Ross OMS-II; Marco Taveras, OMS-II; Rebekah Morrow, PhD VCOM-Louisiana 3
EXAMINING TAU EXPRESSION AND PRESENCE
Context: Endometriosis is a benign gynecological condition characterized by the presence of endometrial-like tissue outside the uterus, often causing chronic pain and infertility. Recent studies have demonstrated an increased risk of developing endometriosis in metabolic conditions derived from insulin dysregulation such as Type 1 diabetes, gestational diabetes and metabolic syndrome. Disturbances in the gut-microbiome have been linked to both endometriosis development and metabolic disorders by altering circulating bacterial derived metabolites, yet precise mechanisms remain unknown. Indole-3-propionic acid (IPA), a gut-bacteria-derived metabolite that has functions in other female reproductive tissues and pathologies, has previously been shown to modulate insulin signaling and glucose homeostasis. Objective: The objective of this study is to determine if IPA can modulate insulin signaling endometriotic cells which may ultimately alter numerous cell processes that may contribute to the severity of the disease.
Methods: For these experiments, we utilized the endometriotic epithelial cell line 12Z. To examine the effects of IPA on insulin signaling, cells were cultured for 24 or 72 hours in the presence or absence of 100nM insulin (hyperinsulinemic levels) and either 1uM (physiologic levels) or 5uM (supraphysiological levels) IPA. Alterations in insulin signaling we determined on whole cell lysates by western blot analysis following standard protocols. Results: Western blot analysis demonstrated that IPA was able to modulate a few insulin responsive genes such as pAKT, FOXO1/3, SPARC and PPAR- γ in a time and dosage dependent manner. The majority of the effects were seen at the 72hr timepoint. Other insulin responsive genes such as p-mTOR were not impacted by IPA treatment. Conclusions: These initial experiments reveal that IPA is capable of modulating insulin signaling in endometriotic cells. This offers a new area of investigation in understanding how metabolic disturbances involving hyperinsulinemia and the gut microbiome influence the pathogenesis of endometriosis. Further experiments investigating
how IPA alters insulin-induced changes in endometriosis cell migration, proliferation and extracellular matrix dynamics are underway.
produced during infection that has been shown to be an innate immune effector (Balzcon et. al. 2024, Kent et. al. 2020). The interrelationship of tau and A β suggests that unphosphorylated tau could be implicated in the antimicrobial activity exhibited by A β . Objective: The focus of this experiment is to confirm the presence or lack of tau protein in rat endothelial cells with previously introduced plasmids encoding for a tau knockout (KO), 1N4R tau isomer, and Ser-214-Ala tau mutant which cannot be phosphorylated. Methods: Starting with cell lines previously listed, we plan to conduct RNA isolations, PCRs, and western blots on the tau variants: 1N4R, Ser-214-Ala, and tau KO using an empty vector as the control. This will allow us to quantify the translation and subsequent transcription of tau into a functional protein from the pulmonary vascular endothelial cells (PVEC). Anticipated Results: We expect to find tau mRNA and protein in 1N4R and Ser-214-Ala; however, in the tau KO it should not be present and have similar results to our control, an empty
vector. We hope to confirm that our tau variants are being actively transcribed and subsequently translated into a functional tau protein as expected. Conclusions: This confirmatory experiment hopes to aid in future investigations into the connection between phosphorylated endothelial tau and the downstream effects found in patients following a nosocomial pneumonia infection by ensuring the suitability of these variants for future experimental use.
12
13
2025 Research Recognition Day
Biomedical Research: Section 1
Biomedical Research: Section 1
Christine Palma, BS, OMS-II 1 ; Lydia Ta, BS, OMS-II 1 ; Nicholas Wong, BS, OMS-II 1 ; Lin Kang, PhD 1 ; K. Adam Morrow, PhD 1 ; Melissa EH Lipsmeyer MS, PhD 1 1 VCOM-Louisiana 4 EXPLORING THE PROTECTIVE ROLE OF INDOLE-3-PROPIONIC ACID IN OBESITY-DRIVEN BREAST CANCER: INSIGHTS FROM AN IN VITRO MODEL
Codi Vernace, BS, OMS-III 1 ; Sabeen Wazir, BS, OMS-II 1 ; Douglas Le, BS, MS, OMS-II 1 ; Kasia Michalak, BS MS 1 ; Lin Kang, PhD 1 ; Stephen DiGiuseppe, PhD 1 ; Melissa EH Lipsmeyer, ME, PhD 1 1 VCOM-Louisiana 5 DYSREGULATION OF ADIPOCYTE FUNCTION DURING CORONAVIRUS INFECTION
Background: Breast cancer is the second leading cause of cancer death in the U.S. with a variety of genetic and physiological contributors to its development. Obesity is associated both with a higher risk of breast cancer development, and with worse disease outcomes through several mechanisms including chronic inflammation, insulin resistance, and increased production of estrogen from adipocytes, all of which enhance cell proliferation. Furthermore, obesity-related metabolic changes can influence the composition of the gut microbiome, leading to dysbiosis, which may further affect breast cancer risk and outcomes by exacerbating inflammation, altering the hormonal milieu or through secretion of bacterial-derived metabolites that directly impact breast cancer cell function. Indole-3-propionic acid (IPA) is a gut-microbiome derived metabolite with anti inflammatory, anti-oxidative stress and glucose regulatory properties that is reduced in patients with overweight/obesity and has been shown to have cytostatic properties against breast cancer cells. However, precise mechanisms by which IPA elicits these functions, especially in the context of obesity, remain unknown.
Hypothesis: The goal of this study was to determine if IPA could alter obesity-induced changes of a hormone receptor positive (ESR, PGR and HER2) breast cancer cell line, MCF-7, in an in vitro model of obesity. Methods: For these studies, an in vitro model of obesity was utilized where breast cancer cells are cultured in media conditioned by mature human adipocytes (ACM) to mimic an obesogenic environment. MCF-7 cells were treated with either control, vehicle, ACM, IPA (1uM) or ACM+IPA media for 72 hours. Next Generation RNAsequencing analysis was used to identify novel gene pathways altered by ACM and if IPA could reverse these changes. Genes with a significant p-value (<0.05) and a log-fold greater than 2 or less than 0.5 were selected and analyzed in a public bioinformatic tool. Results: Pathway enrichment of significantly altered genes revealed that ACM induced changes in gene networks related to estrogen and insulin signaling as well as cellular tight junctions and focal adhesions. Addition of IPA to ACM treated cells resulted in reduction of insulin-signaling pathways as well as
progesterone and estrogen receptor mediated signaling. Experiments to confirm these changes via QPCR and western blot analysis are ongoing. Conclusion: Based on the preliminary RNAsequencing analysis, ACM induced gene expression changes in MCF7 cells that enhance cell survival or proliferation, and IPA may be able to mitigate these effects. Further validation and characterization of the function of IPA in breast cancer cells is needed to determine its potential therapeutic potential in the context of obesity and provide a mechanism by which balance of the gut-microbiome offers a protective role against the disease.
Context: SARS-CoV-2, the causative virus for the COVID-19 pandemic, is estimated to have caused almost 7 million deaths worldwide according to the World Health Organization. Many COVID-19 patients have other comorbidities such as obesity or diabetes mellitus and are at higher risk of complications and mortality. Reports have shown that these patients develop uncontrolled hyperglycemia that exacerbate the development of severe COVID-19 partly due to inflammation and insulin resistance. Emerging studies have demonstrated that dysfunctional adipose tissue due to SARS-CoV-2 infection likely contributes to the development of hyperglycemia in these patients. However, the precise molecular mechanisms behind the pathological changes in adipose tissue due to viral infection remains elusive. Objective: This study aims to investigate the molecular mechanisms by which adipocytes alter their physiology and insulin signaling in response to coronavirus infection. Methods: As a safe model of SARS-CoV2 infection, we utilized human coronavirus OC43 (HCoV-OC43) to infect mature adipocytes that
had been differentiated from subcutaneous fibroblast stem cells. Adipocytes were infected with HCoV-OC43 for 72 hours, after which samples were collected for RNA sequencing, quantitative PCR (qPCR) and western blot. analyses. Results: Human adipocytes were successfully infected with HCoV-OC43 after 72 hours as evidenced by the presence of OC43 nucleocapsid and spike proteins by western blot analysis. Further western blot analysis determined that OC43 infection significantly altered expression of genes essential for adipocyte function. Ongoing RNA sequencing analysis aims to provide a comprehensive overview of the physiological pathways disrupted by HCoV-OC43 infection. Conclusions: Our findings demonstrate that the human coronavirus HCoV-OC43 can effectively infect differentiated adipocytes and significantly alter genes related to adipocyte function. These alterations not only suggest a proposed mechanism through which SARS-CoV2 may impact metabolic health but may give reason to the potential exacerbations seen in patients with pre-existing comorbidities such as obesity.
14
15
2025 Research Recognition Day
Biomedical Research: Section 1
Biomedical Research: Section 1
Grace Lee, OMS-II, Berony Geneste, OMS-II, Dalal Dawud, B Pharm; Zakaria Y Abd Elmageed, PhD Department of Biomedical Science, Discipline of Pharmacology, VCOM-Louisiana 7 REPURPOSING ATYPICAL ANTIPSYCHOTICS FOR PROSTATE CANCER THERAPY: EVALUATING THEIR EFFECTS ON CELL PROLIFERATION, MIGRATION, AND ER STRESS INDUCTION
Jedidiah Lim, OMS; Victoria Lucas, OMS; Samreen Shah, OMS; Sherine Thomas, OMS; Meredith Gwin, PhD; Dara W. Frank, PhD; Samir Gauta, MD, PhD; Melissa Lipsmeyer, PhD; Rebekah Morrow, PhD; K. Adam Morrow, PhD; Sarah Voth, PhD 1 VCOM-Louisiana; 2 Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT; 3 Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI; 4 VCOM-Louisiana 6 PSEUDOMONAS INDUCED ARDS PROMOTES COAGULOPATHY SECONDARY TO CYSTATIN-C DEPLETION
Rationale: Pseudomonas aeruginosa is a predominant agent of pneumonia and infection induced acute respiratory distress syndrome (ARDS). Strains responsible for acute infection utilize a type III secretion system (T3SS) to inject toxic exoenzymes directly into host cells. The T3SS toxin expressed by ~ 90% of clinical strains is exoenzyme Y (ExoY). Intoxicated cells are afflicted with tau hyperphosphorylation, cytoskeletal collapse, and the release of pathogenic tau into the extracellular space. In the absence of ExoY-elicited release of cytotoxic tau, endothelial amyloids including amyloid beta (A β ) are cytoprotective, antimicrobial, and contribute to regulation of the coagulation cascade. Cytotoxic tau disrupts the innate function of endothelial amyloids, suppresses antimicrobicity, and may promote infection induced hypercoagulability, a devastating complication of ARDS. Cystatin C (CysC), a ubiquitous cysteine protease inhibitor, plays a pivotal role in amyloid homeostasis. In critically ill patients, blood and urine proteomics indicate that serum CysC is markedly depleted through ARDS-induced inflammatory proteinuria, yet no cause, remedy, or mechanism of this critical care phenomenon has been identified.
Here, we sought to determine whether CysC depletion via inflammatory proteinuria promotes hypercoagulability during ExoY induced pneumonia. Methods: Male and female wildtype C57BL/6J mice (10-12 weeks old) were infected intratracheally with either vehicle or 1 x 105 colony forming units (CFU) of bacteria in 40 µl of PBS. P. aeruginosa strain ExoY+ (secretes only ExoY in host cells) was used for infections. Weight, urine, and PT/INR were collected daily and survival was tracked. At 48 hours post-infection, surviving mice were sacrificed. Urine was collected via cystocentesis, blood was collected via cardiac puncture, and the brain, kidneys, heart, and lungs were fixed for histological analysis. CysC levels of the blood and urine were measured via ELISA. Amyloid and tau levels were assessed via immunoblotting and thioflavin T.
Background & Aim: Repurposing atypical antipsychotics for untreated metastatic castration-resistant prostate cancer (mCRPC) therapy is an intriguing area of research. Several studies have shown that some antipsychotic drugs, originally developed to treat psychiatric disorders, exhibit anticancer properties. This study aimed to evaluate atypical antipsychotics anticancer properties for treatment of mCRPC by reducing cellular proliferation and migration while inducing endoplasmic reticulum (ER) stress, with fewer side effects compared to first generation antipsychotics. Methods: This study explored the anticancer potential of four atypical antipsychotics: cariprazine, xanomeline, pimavanserin, and iloperidone. Cytotoxic assay was performed to determine the half-maximal inhibitory concentration IC50 values of these drugs in mCRPC cells. Colony formation assay was also used to assess the ability of cells to inhibit clonogenic survival. Transwell migration assay was conducted to evaluate the impact of these agents on cell migration. To elucidate the underlying molecular mechanisms, immunoblotting analysis was conducted to
evaluate pimavanserin’s role in inducing ER stress by quantifying protein markers such as CHOP, BiP, calnexin, PDI, and PERK. Results: We found that MTT assay identified a wide range of IC50 values for the tested antipsychotics, with pimavanserin demonstrating the highest potency in reducing cell viability. Therefore, pimavanserin was selected for further investigation due to its superior efficacy and tolerability profile. Colony formation assay further confirmed pimavanserin’s superior ability to impair clonogenic survival. The transwell migration assay revealed significant inhibition of mCRPC cell migration by all four drugs, with pimavanserin exhibiting the strongest effect. These results highlight pimavanserin as a promising candidate for mCRPC therapy. Western blot analysis will validate whether pimavanserin induces ER stress by upregulating markers such as CHOP, BiP, calnexin, PDI, and PERK, which are critical components of the unfolded protein response. Conclusion: Repurposing atypical antipsychotics offers a promising approach to cancer treatment. Among the tested agents,
pimavanserin displayed the most potent effects in reducing mCRPC cell proliferation, clonogenic survival, migration, and positioning it as a leading candidate for further research. Ongoing studies will focus on elucidating the molecular mechanisms underlying its anticancer activity, particularly its capacity to induce ER stress. These results support the potential development of pimavanserin for preclinical and clinical trials. Keywords: mCRPC, atypical antipsychotic agents, cel proliferation, colony formation, migration
Results: TBD
Conclusions: TBD
16
17
2025 Research Recognition Day
Biomedical Research: Section 1
Biomedical Research: Section 1
Hamza Mehmood, OMS-II 1 ; Ishrar Shaid, OMS-II 1 ; Mohammed Almosa, OMS-II 1 ; Oliver McGhee 2 ; Hassan Ebrahim, PhD 1 ; Khalid El Sayed, PhD 2 1 VCOM-Louisiana; 2 ULM College of Pharmacy 9 THE DIBENZOFURAN LICHEN ACID: A NOVEL SCAFFOLD FOR THE CONTROL OF TRIPLE-NEGATIVE BREAST MALIGNANCIES
David Kang, OMS-II; Vishveshvar Ramkumar, OMS-II; Jedidiah Lim, OMS-II; Aditi Patel, OMS-II; Dalal Dawud, Pharm D; Hassan Ebrahim, PhD; Zakaria Abd Elmageed, PhD VCOM-Louisiana 8 ROSEMARY: A TIMELESS HERB WITH A PROMISING ANTI-BREAST CANCER PROPERTIES
Context: Breast cancer (BC) is the most common cancer diagnosed among American women. Approximately 1 in 8 American women will be diagnosed with BC in her lifetime and 1 in 43 will die from the disease complications. BC encompasses various disease subtypes with different biological, clinical and prognostic characteristics. Despite significant progress over the past several decades in reducing BC mortality, multiple disease challenges remain unmet including the development of resistance to the current standard therapy, intolerability and high rate of recurrence. Dietary phytochemicals have been proven for their potential health benefits, including antioxidant, anti-inflammatory, antiviral, anticancer, and immunomodulation. Phytochemicals harbor an amalgam of diverse biomolecules with the potential capacity to modulate the intricate biological systems in an efficient and selective manner. Objectives and/ or Hypothesis: Previous studies supported the significant role of rosemary-derived phytochemicals in modulating various human diseases, including cancer. Herein we aimed at identifying bioactive phytochemicals in rosemary extract that are more likely to
mediate the anti-BC activity of the herb and investigating their molecular targets.
the polyphenolic rosmarinic acid (RA). RA significantly suppressed proliferation of human BC cell line panel at low µM doses, with MDA MB-231 being the most sensitive BC cell line with IC50 value of 33.2 µM. Additionally, RA inhibited the migration of MDA-MB-231 cells in the scratch assay at 21.7 µM, while effectively suppressed the cell colonization at 11.5 µM. The computational modeling predicted the MDA-MB-231 overexpressed receptor tyrosine kinase c-Met as a potential target for RA in these cells. Western blot analyses successfully validated the target, where RA downregulated the phosphorylated c-Met in a concentration dependent manner. Conclusion: Dietary phytochemicals will remain an innovative resource for the discovery of anticancer biomolecules. The rosemary-derived RA is a novel c-Met tyrosine kinase inhibitor with potential anti-BC activity against c-Met overexpressing triple-negative BC cells. Further preclinical validation is highly warranted to promote the polyphenolic RA to the drug lead rank.
Context: Breast cancer (BC) remains the most commonly diagnosed cancer in American women, with over 40,000 deaths projected in 2025. BC encompasses various disease subtypes with distinct biological, clinical and prognostic characteristics. Among these, triple negative breast cancer (TNBC) is characterized by the lack of positive staining for the estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor 2 (HER2). This lack of targetable receptors renders TNBC inherently resistant to the current BC- targeted therapies, resulting in poorer recurrence-free and overall survival rates compared to other BC subtypes. The receptor tyrosine kinase c-Met, a membrane-bound receptor activated by hepatocyte growth factor (HGF), plays a crucial role in promoting cell proliferation, survival, motility, and angiogenesis. c-Met is frequently overexpressed or dysregulated in TNBC, making it a compelling therapeutic target. Lichens, a symbiotic association between a fungus (mycobiont) and a photosynthetic partner (photobiont, typically alga or cyanobacterium), serve as a unique resource of the discovery of bioactive compounds.
Objectives and/or Hypothesis: Our previous studies identified the dibenzofuran usnic acid (UA) from Louisiana-grown lichens and developed more than 50 chemically optimized analogs with notable anticancer activity, selectively targeting TNBC cells. This study aims to further investigate the molecular targets of these analogs that are most likely mediating their anticancer mechanism. Methods: Analog U26 was synthesized using Claisen-Schmidt carbon-carbon coupling, and its identity was confirmed by NMR and mass spectrometry. Phenotypic screening assays, including cell proliferation, migration and colonization, were implemented to evaluate the anticancer effect of U26 on TNBC cells. Additionally, immunoblotting was utilized to investigate the proteomic profile of TNBC cells treated with U26. Results: U26 was successfully synthesized and demonstrated significant suppression of TNBC cell proliferation, migration and colonization at sub-micromolar levels. Western blot analyses revealed a dose dependent downregulation of c-Met receptor tyrosine kinase in U26-terated TNBC cells.
Conclusion: Natural products and their analogs continue to serve as valuable resources for discovering novel anticancer biomolecules. The lichen-derived dibenzofuran usnic acid analogs, such as U26, are potential c-Met modulators for TNBC treatment. Further preclinical studies are necessary to validate U26 as a lead compound for therapeutic development.
Methods: Polarity-based fractionation of the rosemary extract was accomplished by liquid liquid extraction. The pooled fractions were then tested against multiple human BC cells and bioactive sub-fractions were subjected to extensive chromatographic purification to yield bioactive molecules, which were subsequently evaluated in different cell-free and cell based bioassays including cell proliferation, migration and colonization. The potential macromolecular targets of these purified phytochemicals were disclosed utilizing computational and Western blot analyses. Results: The partition extraction of rosemary yielded four fractions: hexane, ethyl acetate, n-butanol and water. The butanol fraction showed the most promising anticancer cancer activity against MCF-7, MDA-MB-231, MDA MB-468 and BT-474 human BC cell lines in proliferation assays with IC50 values of 104.2, 43.7, 45.9, and 91.6 µg/mL, respectively. Further chromatographic separation identified different phytochemicals, including
18
19
2025 Research Recognition Day
Biomedical Research: Section 1
Biomedical Research: Section 1
Mohammed Almosa, OMS-II; Hamza Mehmood, OMS-II; Ishrar Shaid, OMS-II; John Woeste, OMS-II; Christian Caper, OMS-II; Dalal Dawud, Pharm D; Zakaria Abd Elmageed, PhD; Hassan Ebrahim, PhD VCOM-Louisiana 10 THE SHRUBBY SOPHORA SUPPRESSES THE ONCOGENESIS OF HUMAN BREAST CANCER CELLS
Hunter Santogrossi, DO 2 ; Ryan Vergara, DO 2 ; Keith Jackson, PhD 2 ; Dinesh Aryal, PhD 2 1 VCOM-Louisiana; 2 ULM, College of Pharmacy 11 EFFECT OF RENAL HEME OXYGENASE ON RENAL VASCULAR BLOOD FLOW DURING CHRONIC ACIDOSIS CONDITIONS IN RATS
Context: wNatural products (NPs), also known as secondary metabolites, constitute a unique resource for the discovery of validated hits, lead compounds and drug candidates. NPs are characterized by enormous scaffold diversity and structure complexity when compared to synthetic molecules making them ideals templates for early drug discovery. Furthermore, their structural three dimensionality, rigidity, and chirality empower NPs to interrogate the complex biological systems in a selective and efficient manner. Shrubby Sophora is a flowering plant belonging to family Fabaceae and is native to China. It harbors NPs of diverse chemical classes including alkaloids, flavonoids, isoflavonoids, and prenylated phenolics. Breast cancer (BC) is the most common diagnosed cancer and the second leading cause of death among women worldwide. In the US, statistics approximate that 1 in 8 American women will be diagnosed with BC during lifetime, and 1 in 43 will die from the advanced BC, highlighting the significant impact of the disease. Objectives and/or Hypothesis: Previous studies demonstrated the significant role of Sophora-derived phytochemicals in modulating
various human diseases, including cancer. Herein we aimed at identifying bioactive phytochemicals in the shrubby Sophora extract that are most likely mediating the anti BC activity and investigating their potential macromolecular targets. Polarity-based fractionation of the shrubby Sophora extract was executed by partition extraction technique. Fractions were tested against multiple human BC cells and to guide the efforts towards identifying biomolecules that most likely mediating the anticancer effect of Sophora. Cell proliferation, apoptosis, and cell cycle analysis were used to characterize the anticancer effect of the purified compounds. Computational molecular modeling and enzymatic assays were utilized to uncover the mechanistic target. Results: Shrubby Sophora extract significantly suppressed the proliferation of MDA-MB-468, MDA-MB-231, MCF-7, and BT-474 human BC cells in a concentration-dependent manner at low µg/mL concentration range. Additionally, the bioassay-guided fractionation of Sophora extract have led to the discovery of a pterocarpan flavonoid with potent anticancer activity at low µM level (IC50 values of 17.6,
19.1, 14.7 and 16.5 µM against the BC cell panel, respectively). Furthermore, it induced cell apoptosis and cell cycle arrest at G2/M phase. Computational molecular modeling suggested DNA topoisomerases as potential macromolecular targets, which was validated by DNA relaxation assays. Conclusion: NPs will stand as innovative resources for the discovery of anticancer hits. The bioassay-guided fractionation of shrubby Sophora extract have led to the discovery of a pterocarpan flavonoid as a DNA topoisomerases inhibitor with potential anticancer activity against human BC cells. Future preclinical studies are encouraged to validate the bioactivity of this novel herb.
Background: Chronic metabolic acidosis (CMA) characterized by prolonged systemic acidemia have been associated with various pathological conditions. Hypertension, as an outcome of prolonged acidosis, has been recently observed in experimental animals. It is documented that heme oxygenase-1 (HO-1), influences renal hemodynamics by affecting vascular tone and blood flow, potentially contributing to the regulation of blood pressure. We recently reported the renal HO-1 levels rise during chronic acidosis. The cellular and molecular events mediating the effects of acidosis on vascular homeostasis are incompletely understood. In the present study, we investigated the effect of HO-1 on renal vascular blood flow as well as the role of renal HO-1 on renal oxidative stress during chronically acidotic conditions. Objectives: (i) To examine renal blood flow during chronic acidosis condition to determine the potential renovascular hypertension, (ii) To measure the renal oxidative stress mediated by renal HO-1 during chronic acidosis conditions.
(I. Control, II. CMA, III. CMA+DALA, IV. CMA+Fasudil, V. CMA+ZnPP). 0.28M ammonium chloride induced chronic acidosis model is implemented for the latter 4 groups along with respective inducer/inhibitor treatments (once a week), for the period of 8 weeks. At the end of the experiment, renal arteries of all animals were examined for renal blood flow using Laser Doppler Flowmeter. Arterial walls were observed and quantified via imaging. ELISA was performed to measure the eNOS levels in the whole blood samples collected from each animal. The microdialysis samples taken from kidney interstitium of each animal were analyzed for their superoxide dismutase (SOD) levels and HO-1 levels. All animals used in this work were approved by IACUC at University of Louisiana at Monroe (Protocol # 23 SEPT DA_KEJ 01). Results: HO-1 levels in the renal interstitium of Group II were significantly elevated vs control group. The man arterial pressure (MAP) rises significantly in the CMA group though, as expected. The renal blood flow in all five groups weren’t significantly changed. Furthermore, there was no sign of stenosis in all the animals examined. Interestingly, the eNOS levels in CMA
was reduced significantly vs control group. This suggested that the elevated renal HO-1 in CMA group might have reduced the nitric oxide (NO) release in the vasculature which perhaps led to rise in MAP. No changes were seen in eNOS levels in groups: III & V, compared to group II. However, group IV showed a significant increase in eNOS levels vs group II. SOD levels in group II were reduced significantly vs the control group. Surprisingly, ROCK inhibitor (Fasudil) treated group had significantly increased levels of SOD compared to the CMA group. Conclusions: No renal blood flow was impaired in the experimented animals subjected to chronic metabolic acidosis. This suggested, no renovascular hypertension is contributing to the increase in MAP in the CMA group. Renal HO-1 may inhibit the release of eNOS leading to declining nitric oxide to promote the observed hypertension. Rho Kinase (ROCK) played a significant role compared to HO-1 to reduce the renal oxidative stress during chronic metabolic acidosis. Future studies are likely to further define the importance of HO-1 and ROCK synergism if any existed to clarify their independent role in chronic acidosis.
Methods: Male Sprague Dawley rats weighing 150-320g were grouped into 5 groups (n=5).
20
21
2025 Research Recognition Day
Biomedical Research: Section 1
Biomedical Research: Section 1
Jedidiah Lim 1 ; David Kang 1 ; Victoria Lucas 1 ; Samreen Shah 1 ; Sherine Thomas 1 ; Carlie Christ 1 ; Meredith Gwin, PhD 2 ; Samir Gautam, MD, PhD 2 ; Melissa Lipsmeyer, PhD 1 ; Rebekah Morrow, PhD 3 ; K. Adam Morrow, PhD 1 ; Sarah Voth, PhD 1 1 Cell Biology and Physiology, VCOM-Louisiana; 2 Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine; 3 Microbiology and Immunology, VCOM-Louisiana 12 PSEUDOMONAS INDUCED ARDS PROMOTES INTRAPULMONARY MICROTHROMBUS FORMATION AND ACUTE LUNG INJURY SECONDARY TO CYSTATIN-C DEPLETION
Zachary Dickey, OMS-IV; Annie Pham, OMS-III; Krishna Patel, BS; Melissa Lipsmeyer, PhD; Kasia Michalak, MSc; Troy Stevens, PhD; Rebekah Morrow, PhD; K. Adam Morrow, PhD; Sarah Voth, PhD Dept of Cell Biology and Physiology, VCOM; Dept of Physiology & Cell Biology, USA COM; Dept of Microbiology & Immunology, VCOM 13 PSEUDOMONAS AERUGINOSA TOXIN ExoY INHIBITS SIRT1-MEDIATED AUTOPHAGY TO PROMOTE PULMONOGENIC TAUOPATHY
Rationale: Acute respiratory distress syndrome (ARDS) is a critical respiratory condition caused by the accumulation of fluid in the lungs which impairs gas exchange and ultimately leads to respiratory failure. ARDS is commonly caused by infections from microorganisms, most notably Pseudomonas aeruginosa. Strains responsible for acute infection utilize a type III secretion system (T3SS) to inject toxic exoenzymes directly into host cells. Approximately 90% of clinical strains expressed involve exoenzyme Y (ExoY). Intoxicated cells are afflicted with tau hyperphosphorylation, cytoskeletal collapse, and the release of pathogenic tau into the extracellular space. In the absence of ExoY elicited release of cytotoxic tau, endothelial amyloids including amyloid-beta (A β ) are cytoprotective, antimicrobial, and stabilize the endothelial cell microtubule network to establish barrier integrity within the lungs. Cytotoxic tau disrupts the innate function of endothelial amyloids, suppresses antimicrobicity, and collapses the microtubule barrier in the lungs, leading to the dissemination of cytotoxic tau and beta-amyloid into the circulation of other peripheral organs causing end-organ damage. Cystatin C (CysC), a ubiquitous
cysteine protease inhibitor, maintains amyloid homeostasis. In critically ill patients, blood and urine proteomics indicate that serum CysC is markedly depleted through ARDS-induced inflammatory proteinuria, yet no cause, remedy, or mechanism of this critical care phenomenon has been identified. In these studies, we investigated whether ExoY-competent P. aeruginosa infection induces intrapulmonary microthrombus formation and acute lung injury with concomitant loss of serum CysC. Methods: Male and female wildtype C57BL/6J mice (10-12 weeks old) were infected intratracheally with either vehicle or 1 x 105 colony forming units (CFU) of bacteria in 40 µl of PBS. P. aeruginosa strain ExoY+ (secretes only ExoY in host cells) was used for infections. Weight, urine, and PT/INR were collected daily, and survival was tracked. At 48 hours post-infection, surviving mice were sacrificed. Urine was collected via cystocentesis, blood was collected via cardiac puncture, and the brain, kidneys, heart, and lungs were fixed for histological analysis (H&E and Congo Red). Bronchoalveolar lavage fluid was collected, protein concentration measured, and cell
populations determined via Coulter Counter. CysC levels of the blood and urine were measured via ELISA and immunoblotting. Amyloid and tau levels were assessed via immunoblotting and thioflavin T.
Rationale: Survivors of nosocomial pneumonia often suffer an acute-to-chronic disease transition. Despite resolution of active infection, secondary end-organ damage promotes chronic illness and decreased longevity. Accumulating evidence implicates prion like cytotoxic tau elicited from infected lung capillary endothelium as a notable contributor to this phenomenon. Common nosocomial Pseudomonas aeruginosa utilizes a type III secretion system (T3SS) during acute lung infection. Roughly 90% of clinical isolates express the T3SS toxin Exoenzyme Y (ExoY). ExoY induces unregulated cNMP production, tau hyperphosphorylation, cytoskeletal collapse, antimicrobial amyloid loss-of-function, and the generation of prion-like cytotoxic tau. However, the mechanisms responsible remain unclear. Sirtuin 1 (SIRT1) deacetylase is intricately linked to cellular bioenergetics including autophagy, a fundamental means of clearing dysregulated protein. Here, we tested the hypothesis that ExoY intoxication constrains SIRT1-mediated autophagy to promote the production and release of cytotoxic tau.
cells were serum starved and infected with isogenic strains of P. aeruginosa at an MOI of 20:1 in HBSS with/without the clinical SIRT1 agonist SRT2104. Strains included a mutant incapable of generating cNMPs (ExoYK81M) in host cytosol and a mutant that injects only cNMP-competent ExoY (ExoY+) into host cells. At 4 hours post-infection, whole-cell lysates, RNA, and supernatants were collected. Supernatants were filter-sterilized, boiled, and iced. Lysates were immunoblotted for SIRT1, Tau (TNT-1 antibody), and LC3, and supernatants probed for tau. Transcript levels of SIRT1, Tau, and LC3 were measured using RT-qPCR and immunofluorescence studies determined the cellular localization of SIRT1, Tau, and LC3. Antimicrobial studies (e.g., Kirby Bauer) and cytotoxicity assays (e.g., resazurin) were utilized to determine the downstream impact of cell supernatants. Results: ExoY intoxication significantly diminished protein and transcript levels of SIRT1 while abolishing LC3 in PMVECs as compared controls. The supernatant-to-lysate ratio of tau protein in ExoY+ infected cells was more than double that of controls. ExoY-intoxicated
cells treated with SRT2104 exhibited complete rescue of LC3 and SIRT1 levels, robust LC3-I to LC3-II conversion, and tau was ablated in the supernatant fraction. Immunofluorescence revealed SRT2104 treatment of ExoY-intoxicated cells promotes tau co-localization with LC3 equivalent to vehicle. Supernatants from ExoY-intoxicated cells were cytotoxic whereas SRT2104 treatment produced ExoY-generated cell supernatants with antimicrobicity equivalent to controls.
Results: TBD
Conclusions: TBD
Methods: Pulmonary microvascular endothelial
22
23
2025 Research Recognition Day
Made with FlippingBook Ebook Creator