We use cookies to distinguish you from other users and to provide you with a better experience on our websites. Close this message to accept cookies or find out how to manage your cookie settings.
This journal utilises an Online Peer Review Service (OPRS) for submissions. By clicking "Continue" you will be taken to our partner site
https://mc.manuscriptcentral.com/jcts.
Please be aware that your Cambridge account is not valid for this OPRS and registration is required. We strongly advise you to read all "Author instructions" in the "Journal information" area prior to submitting.
To save this undefined to your undefined account, please select one or more formats and confirm that you agree to abide by our usage policies. If this is the first time you used this feature, you will be asked to authorise Cambridge Core to connect with your undefined account.
Find out more about saving content to .
To send this article to your Kindle, first ensure no-reply@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about sending to your Kindle.
Find out more about saving to your Kindle.
Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.
Objectives/Goals: Neuromodulation strategies like transcranial magnetic stimulation (TMS) can target specific neural circuits underlying particular psychiatric symptoms, potentially 1) enhancing understanding of mechanisms of illness and recovery and 2) acting as novel therapeutics. These feasibility studies lay foundation for a study of major depression. Methods/Study Population: Four healthy volunteers completed structural and functional MRI (fMRI). fMRI included a trait-adjective task, a negative self-referential processing task known to activate VMPFC, which is known to be abnormal in major depression. During the task, participants respond on a task pad whether they feel that each of a series of displayed adjectives (positive, negative, or neutral) applies to them. Three participants then participated in a simulated image-guided TMS session using their MRI data to target their VMPFC. Three-dimensional tracking of the participant’s head and the TMS coil was used to position the coil for peak stimulation of the targeted brain region. Results/Anticipated Results: Our team collected quality neural and behavioral data on the fMRI task; participants reported a tolerable experience. Simulated neuronavigated TMS showed feasibility and tolerability of positioning the device to stimulate VMPFC. The fMRI task activated the VMPFC as predicted. The MRI and TMS protocols were replicable and tolerable. These procedures can now be used experimentally by our team with confidence to test our hypothesis that targeting the VMPFC within the brain’s default-mode network may normalize aberrant VMPFC activity seen in major depression, thereby improving excessive negative self-referential processing. Discussion/Significance of Impact: This project lays essential groundwork for my K12 project, “Targeting Negative-Self Referential Processing in Depression with TMS,” a longitudinal neuroimaging and behavioral study using these methods in the study population of people with major depression.
Objectives/Goals: Increased follicle-stimulating hormone (FSH) is linked to declines in ovarian and metabolic function in older women. Obesity is both a manifestation and a driver of aging pathologies. In animal models, FSH and insulin resistance (IR) were reduced after 6 mos. of a nutritional therapeutic (GLYLO). Our goal was to translate preclinical evidence to humans. Methods/Study Population: An integrated, precision medicine approach identified a unique phenotype of aging-related debility relative to older females. A non-comparer pilot study was conducted to translate GLYLO preclinical findings to postmenopausal women with obesity (n = 85; >55 years; body mass index [BMI] = 35.0±4.35; range: 30.3–42.8). Participants meeting the inclusion and exclusion criteria (n = 13) were enrolled and received two capsules of GLYLO (vitamins and natural products) daily for 6 mos. Assessments for FSH, estradiol (E2), IR (homeostatic model [HOMA-IR]), total cholesterol (TC), low- (LDL), high-density lipoproteins (HDL), safety biomarkers (e.g., red cell distribution width [RDW%], mean corpuscular volume [MCV]), and depression (Center for Epidemiologic Studies Depression Scale) were conducted prior to and after 6 mos. Results/Anticipated Results: Mixed-effect models with intent-to-treat analysis were applied to compare outcomes prior to (n = 13) and following (n = 7) the intervention. Significant reductions in FSH were observed (-13.1 [2.47] ∆/SD; p = 0.002)) following the 6-month intervention. Interestingly, BMI, E2 (p = 0.412), HOMA-IR (p = 0.885), TC (p = 0.363), and LDL (p = 0.145) were unchanged, while HDL decreased significantly (-9.7 [3.82] ∆/SD; p = 0.044). Other biomarkers, RDW% (-0.2 [0.05] ∆/SD; p = 0.009) and MCV (-2.3 [0.33] ∆/SD; p = 0004), were significantly reduced. All other safety parameters were not altered. Six participants reported mild to moderate adverse events (acid indigestion) and were lost to follow-up. Depression scores significantly increased (+4.0 [0.75] ∆/SD; p = 0.002)). Results were similar with and without intent to treat analysis. Discussion/Significance of Impact: Decreased FSH, but not IR, was observed following six months of GLYLO in postmenopausal women with obesity. Significant alterations in HDL, depression, RDW%, and MCV warrant further investigation. Findings are limited by the small sample size and loss to follow-up. Randomized, controlled trials are needed to confirm these results.
Objectives/Goals: Large-scale tumor sequencing efforts have led to annotations of novel cancer hotspot mutations that may underlie driver or cooperative function. We have sought to define the molecular consequences of such hotspots associated with pediatric DICER1 syndrome cancers, with the ultimate goal of revealing novel targets that may inform new standards of care. Methods/Study Population: We have performed genomic analysis to identify tumor types (in TCGA and MSK-IMPACT patient data) for which mutations in the Dicer1 gene (encoding Dicer protein) emerge as the dominant signature of driver function. As Dicer is a critical RNA processing factor responsible for the generation of microRNAs, which are posttranscriptional gene regulatory molecules, we have modeled these mutations in human embryonic stem cells in order to study the direct effects on miRNAs and their target genes in an isogenic background. In addition to providing the required setting for unambiguous attribution of function to specific mutations, clonal human ES cells offer an opportunity for modeling of both developmental and cancer requirements associated with altered Dicer function. Results/Anticipated Results: Through generation of genomics and functional datasets from matched genotypes in Dicer mutated human ES cells, we have identified specific alterations in miRNAs and their effects on target genes. Unexpectedly, we found direct evidence for both loss of function and gain of function attributable to Dicer mutations. In addition, through integrated analysis of genomic data from tumor sequencing datasets and our human ES cell models, we have identified potential miRNA and target gene alterations that underlie tumorigenic potential, nominating gene candidates for targeted therapy in DICER1 syndrome. Direct mouse modeling of such candidate gene targets has revealed evidence for driver function of identified miRNA and their targets. Discussion/Significance of Impact: DICER1 syndrome cancers comprise a wide variety of rare pediatric tumor types. Presently, we still lack an effective standard of care. Furthermore, the previous lack of molecular profiling precluded targeted therapy opportunities. Our precise knock-in modeling of Dicer hotspots and deep profiling of relevant tumors now provide candidate targets.
Objectives/Goals: Cutaneous lupus erythematosus (CLE) is an inflammatory skin manifestation of lupus. CLE lesions are frequently colonized by Staphylococcus aureus, a microbe known to promote IFN production and inflammation. Here, we investigate whether type I IFN and inflammatory gene signatures in CLE lesions can be modulated with a topical antibiotic treatment. Methods/Study Population: SLE patients with active CLE lesions (n = 12) were recruited and randomized into a week of topical treatment with either 2% mupirocin or petroleum jelly vehicle. Paired samples were collected before and after 7 days of treatment to assess microbial lesional skin responses. Microbial samples from nares and lesional skin were used to determine baseline and posttreatment Staphylococcus abundance and microbial community profiles by 16S rRNA gene sequencing. Inflammatory responses were evaluated by bulk RNA sequencing of lesional skin biopsies. Immunophenotyping of CLE lesions was performed using CIBERSORTx to deconvolute the RNA-seq data into predicted cell populations impacted by treatment. Results/Anticipated Results: We identified 173 differentially expressed genes in CLE lesions after topical mupirocin treatment. Mupirocin treatment decreased the abundance of Staphylococcus associated with CLE lesions without altering the overall diversity of the skin microbiota relative to vehicle. Decreased lesional Staphylococcus burden correlated with decreased IFN pathway signaling and inflammatory gene expression and increased barrier dysfunction. Interestingly, mupirocin treatment lowered skin monocyte levels, and this mupirocin-associated depletion of monocytes correlated with decreased inflammatory gene expression. Discussion/Significance of Impact: Mupirocin treatment decreased lesional Staphylococcus burden and this correlated with decreased IFN signaling and inflammatory gene expression. This study suggests a topical antibiotic could be employed to decrease lupus skin inflammation and type I IFN responses by reducing Staphylococcus colonization.
Objectives/Goals: Our main objective was to compare 5-year survival and organ function between patients with sickle cell disease (SCD) who underwent hematopoietic cell transplant (HCT) and those who did not undergo HCT. We hypothesized that organ function would be improved in those with SCD who underwent HCT when compared to those who remained on standard therapy. Methods/Study Population: This IRB-approved, retrospective study includes patients with SCD treated at Children’s Healthcare of Atlanta. Cases underwent HCT between 2010 and 2016. They were randomly matched with 2 patients with SCD who did not undergo HCT. Match criteria included age, sex, disease genotype, and disease severity, which was determined by the number of hospitalizations in the 5 years pre-HCT, prior intensive care unit admission, and prior chronic transfusion therapy. Data extracted included SCD treatment, hospitalizations, emergency department visits, and organ function pre-HCT and 1-, 2-, 3-, and 5-years post-HCT. Organ-specific outcomes and overall survival were compared between the two groups using cumulative incidence curves and Kaplan–Meier analyses. Normal FEV1 and FVC in this analysis were >80% predicted. Results/Anticipated Results: Thirty-seven cases who had undergone HCT were matched with 74 controls who continued with standard medical therapy. The median age was 8 years for both groups and 59% were females. The median disease severity score was 2 in both groups. At baseline, 70.3% of the HCT group completed pulmonary function tests (PFTs) compared to 35.1% of the non-HCT group. Of these, 73% in both groups had a normal FEV1. In terms of FVC, 57.7% of HCT patients and 76.9% of non-HCT patients had a normal FVC pre-HCT. At 5 years post-HCT, 56.8% of the HCT group had PFTs completed compared to 21.6% of the non-HCT group. Among these, 85.7% in the HCT group had a normal FEV1 compared to 75% in the non-HCT group, while 90.6% had a normal FVC in the HCT group compared to 75% in the non-HCT group. Two of 37 in the HCT group and 1 of 74 in the non-HCT group died (p = 0.21). Discussion/Significance of Impact: Our data suggest that post-HCT, the proportion of patients falling in the normal range for FEV1 and FVC increases. This increase is not seen in the non-HCT group, indicating that HCT may improve this organ function. There was no difference in survival between the groups, indicating the risk of HCT mortality may not be greater than the risk of living with SCD.
Objectives/Goals: As life expectancy increases in people with HIV, neurocognitive impairment is becoming more common, and women with HIV (WWH) are disproportionately impacted. This work investigated mitochondrial function and oxidative stress in WWH in order to understand the relationship between mitochondrial function and cognition in future studies. Methods/Study Population: Peripheral blood mononuclear cells were isolated from virally suppressed WWH (n = 64) and underwent the Seahorse Cell Mito Stress test to assess different realms of mitochondrial function. Cells were then lysed for direct DNA extraction, and quantitative PCR was performed to understand mitochondrial DNA expression (mtDNA) levels as a measure of oxidative stress. A series of simple linear regressions was then conducted to understand the relationships between mitochondrial function and mtDNA content. Future work will expand this analysis to investigate associations between demographic dynamics, such as trauma history, and mitochondrial function, as well as to understand the relationships between mitochondrial function and cognitive outcomes in WWH. Results/Anticipated Results: In our cross-sectional analysis of mitochondrial dynamics in WWH, we found a significant association between maximum mitochondrial respiration ability and mtDNA content, with greater mtDNA expression associated with increased levels of maximum respiration following stimulation. There was no association between basal respiration and levels of oxidative stress. There was also a significant variation in mitochondrial function in our participants, indicating that future analyses to investigate the source of that variation are warranted. The work presented here sheds light on mitochondrial dynamics in WWH and will be the basis for future studies that will investigate how demographic dynamics may be associated with mitochondrial function, as well as how mitochondrial dynamics may predict cognitive outcomes. Discussion/Significance of Impact: There is significant variation in mitochondrial function in WWH. More analysis is needed to understand what may be associated with these variations, including an investigation of both clinical factors as well as cognitive outcomes. This analysis will inform directions for future mechanistic work aimed at mitigating adverse cognitive outcomes in WWH.
Objectives/Goals: Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease with a mean survival of only 11 months even with the most advanced treatment to date. The desmoplastic microenvironment of PDA is thought to play a critical role in therapy resistance. One pathway that might be responsible for resistance to immunotherapy is the CXCR4-CXCL12 axis. Methods/Study Population: In this study, we propose to evaluate the effect of CXCR4-CXCL12 inhibition on dual checkpoint inhibition in KPC mouse model of PDAC and patient-derived explants. PDAC mouse models are made with pancreatic cancer cells driven by loss of TP53 and activation of KRAS. These models are treated with PD1 inhibitor Balstilimab and an FC-modified CTLA4 Botensilimab with or without CXCR4 inhibitor BL8040. In addition, we make explants of patient tumors along with their tumors and autologous peripheral blood mononuclear cells and this model is similarly challenged with BOT/BAL and BL8040. Using immunofluorescence and flow cytometry, we quantify and evaluate the spatial relationships between different cell populations. Most notably, we evaluate the relative abundance of CD8+ T cells in control and treated conditions. Results/Anticipated Results: We expect the inhibition of CXCR4-CXCL12 axis, along with two new potent checkpoint blockers, will lead to infiltration of CD8+ T cells in both the mouse and human PDAC models. We also expect this to translate into more tumor cell killing as demonstrated by Caspase activities and tumor shrinkage. Discussion/Significance of Impact: If our hypothesis is proven in both mouse and human PDAC models, this study will serve as a basis for a phase I/II clinical trial testing this combination of drug.
Objectives/Goals: Osteoarthritis (OA) is a multifactorial disease where sustained gut inflammation is a continued source of inflammatory mediators driving degenerative processes in joints. The goal was to use spontaneous equine model to compare fecal and leukocyte microbiome and correlation to transcriptome in OA. Methods/Study Population: Seventy-six horses (31 OA, 45 controls) were enrolled by population-based sampling. Feces and peripheral blood mononuclear cells (PBMC) were collected. Horses were determined to have OA by clinical and radiographic evidence. Horses were excluded if they received medications or joint injections within two months. Fecal and circulating leukocyte bacterial microbial 16s-seq was performed. Bulk RNAseq of PBMC was performed by the Illumina platform. Gene expression data were mapped to the equine genome, and differential expression analysis was performed with DESeq2. Qiime2 was used for microbial analysis. Enrichment analysis was performed with a cluster profiler. Correlation analyses were performed between the datasets. Results/Anticipated Results: Beta and alpha microbial diversity differed in feces and PBMC of OA vs. healthy horses. Horses with OA had an increased Firmicutes to Bacteroidetes ratio compared with controls. The fecal microbiome of OA horses had significantly higher amounts of Firmicutes Oribacterium (q Discussion/Significance of Impact: These data suggest that altered microbiome and PBMC gene expression are associated with naturally occurring OA in the translational equine model. While Oribacterium has been detected in humans with rheumatoid arthritis, its role in OA warrants further proteomic and metabolomic profiling.
Objectives/Goals: Hearing loss (HL) can result from environmental and genetic factors. Some genetic variants may be more prevalent in populations living in geographic or cultural isolation. This study explores the genetic variants associated with HL in Puerto Rico and correlates these with auditory and balance disorders to uncover novel variants. Methods/Study Population: After obtaining individual informed consent and assent for a minor when applicable, we will collect clinical audiological data and biological samples (n = 600) from families across Puerto Rico with a history of severe to profound HL. Genomic DNA will be extracted, and exome and mitochondrial genome sequencing will be conducted to identify causal variants in genes associated with HL. The study will assess the prevalence of both novel and reported variants in genes associated with HL and investigate founder variants in the Puerto Rican population. Involvement of genes so far not associated with HL will also be considered when a genetic diagnosis cannot be established. Auditory phenotypes will be correlated with genetic findings, allowing for a comprehensive analysis of genetic contributions to HL in this population. Results/Anticipated Results: This research will advance understanding of the genetic causes of HL in Puerto Rico, leading to more accurate diagnoses, personalized treatment options, and the discovery of novel genes associated with HL. It will also serve as an evidence-based reference to analyze the adequacy of current neonatal hearing screening protocols in PR. Recruitment and sample collection have begun, and we expect our findings to uncover population-specific variants. These results will provide a foundation for further genetic studies aiming at identifying the causes of HL in Puerto Ricans regardless of age of onset. Discussion/Significance of Impact: This study will enhance our understanding of hereditary HL and serve as a basis for developing population-specific diagnostic tools and interventions, particularly in the Puerto Rican population. The research will support future genetic studies and address health disparities in HL in the island.
Objectives/Goals: The study’s goal is to investigate the role of PPAR-α on regulating blood pressure, glomerular filtration rate (GFR), renal inflammation, and renal sodium reabsorption in mice on a 4% high-salt diet. Methods/Study Population: GFR, systolic blood pressure (SBP), inflammatory biomarkers (KIM-1, TIMP2, NGAL, MCP-1, TNF-α, IL-6, IL-10, and IL-17), and renal sodium transporter expression (NKA, NHE3, NKCC2, NCC, ENaC, Aqp-2, and NHERF1) were measured in PPAR-α KO mice and wild-type controls treated with a 4% high-salt (HS) diet. Male C57BL6, B129S1, and PPAR-α KO mice (12 weeks old) will be treated with 4% HS diet for 28 days. Systolic blood pressure is measured by tail cuff. GFR is measured by transdermal FITC-Inulin radioactive fluorescence. Inflammatory biomarkers will be measured by cytokine array and western blot. Sodium transporter expression will be measured by western blot. Results/Anticipated Results: Baseline SBP was 146 ± 31 mmHg (C57), 140 ± 24 mmHg (B129), and 153 ± 23 mmHg (KO). After 21 days of normal (control diet) or treatment (HS diet), control systolic pressures were 139 ± 18 mmHg (C57), 107 ± 23 mmHg (B129) and 147 ± 34 mmHg (KO), while HS systolic pressures were 166 ± 23 mmHg (C57) and 119 ± 34 mmHg (B129). We are collecting blood pressure for the KO HS group. Baseline GFR was 1194 ± 140 µL/min/g (C57), 1167 ± 279 µL/min/g (B129), and 1191 ± 157 µL/min/g (KO). Discussion/Significance of Impact: We hypothesize significantly higher SBP, inflammatory marker expression, and renal sodium transporter expression in KO and B129 mice on a HS diet. We predict that PPAR-α expression in the kidney will be higher in C57 compared to B129. We predict that PPAR-α activity plays a vital role in reducing high-salt-induced hypertension and inflammatory markers.
Objectives/Goals: We hypothesized that the bulk transcriptomic profiling of blood collected from within the ischemic vasculature during an acute ischemic stroke with large vessel occlusion (LVO) will contain unique biomarkers that are different from the peripheral circulation and may provide much-needed insight into the underlying pathogenesis of LVO in humans. Methods/Study Population: The transcriptomic biomarkers of Inflammation in Large Vessel Ischemic Stroke pilot study prospectively enrolled patients ≥ 18 years of age with an anterior circulation LVO, treated with endovascular thrombectomy (EVT). Two periprocedural arterial blood samples were obtained (DNA/RNA Shield™ tubes, Zymo Research); 1) proximal to the thrombus, from the internal carotid artery and 2) immediately downstream from the thrombus, by puncturing through the thrombus with the microcatheter. Bulk RNA sequencing was performed and differential gene expression was identified using the Wilcoxon signed rank test for paired data, adjusting for age, sex, use of thrombolytics, last known well to EVT, and thrombolysis in cerebral infarction score. Bioinformatic pathway analyses were computed using MCODE and reactome. Results/Anticipated Results: From May to October 2022, 20 patients were screened and 13 were enrolled (median age 68 [SD 10.1], 47% male, 100% white). A total of 608 differentially expressed genes were found to be significant (p-value) Discussion/Significance of Impact: These results provide evidence of significant gene expression changes occurring within the ischemic vasculature of the brain during LVO, which may correlate with larger ischemic infarct volumes and worse functional outcomes at 90 days. Future studies with larger sample sizes are supported by this work.
Objectives/Goals: This study aims to first understand the expression of the L-type amino acid transporter, Slc7a5, in demyelinated plaques in postmortem multiple sclerosis (MS) CNS tissue. It also seeks to understand the effect of a novel inhibitor of Slc7a5 on remyelination in mice with experimental autoimmune encephalomyelitis. Methods/Study Population: Using single-cell RNA sequencing (scRNA-seq), we will examine the expression of Slc7a5 in demyelinated plaques in postmortem CNS tissue of patients with MS compared to non-lesioned regions (n = 3/group). Using visually evoked potential (VEP) on mice with experimental autoimmune encephalomyelitis (EAE), we will determine the ability of the Slc7a5 allosteric inhibitor OKY-034 to promote remyelination compared to EAE-only controls (n = 10/group). Lastly, we will use spatial transcriptomics with scRNA-seq to map transcriptional activity within different populations of cells to determine how OKY-034 changes gene expression in specific cell types compared to EAE-only controls (n = 3/group). Results/Anticipated Results: A conditional knockout of Slc7a5 showed that microglial activation and oligodendrocyte differentiation were affected in demyelinated lesions. This suggests that it plays a role in numerous cell types in active demyelinated plaques, which is what we expect to find from our scRNA-seq data in post-mortem CNS tissue of patients with MS. Measuring VEP is a noninvasive way to measure remyelination in both clinical and research settings. OKY-034 increases oligodendrocyte differentiation suggesting remyelination, so we expect that administration of OKY-034 in mice with EAE will lead to restored VEP compared to control and EAE-only mice. Lastly, because OKY-034 reduces inflammation, we expect to see a decrease in gene expression for genes involved in an immune response. Discussion/Significance of Impact: Completion of this study will lead to understanding what the effect the allosteric Slc7a5 inhibitor OKY-034 has on remyelination and whether it may serve as a novel therapeutic drug that can be administered orally for the treatment of MS. This could lead to its further development as a treatment for progressive MS.
Objectives/Goals: Personalized cancer therapy based on genomic testing is advancing patient care. Genomic alterations in fibroblast growth factor receptor (FGFR) predict response to FGFR inhibitors; however, the role of RNA expression and protein activation is not known. We propose to examine the phospho-proteomic signature in FGFR-altered cancers to identify new candidates for FGFR-targeted therapies. Methods/Study Population: In our preliminary study, we have curated a cohort of FGFR2 mutants (13 FGFR2-fusions and 4 FGFR2 point mutations) with known clinical outcomes to FGFR inhibitors and 8 FGFR2 wild-type (WT) cholangiocarcinoma tumor samples to investigate the phospho-proteomic fingerprint using a clinical grade reverse phase protein array (RPPA). RPPAs are high throughput quantitative antibody-based proteomics assays that can quantify hundreds of proteins in thousands of patient tissues providing a high degree of sensitivity through laser tumor microdissection (LCM). We have selected proteins in the FGFR signaling pathway including FGFR2, AKT, ERK1.2, STAT1/3, FRS2, and PLCg to define the range of phospho-proteomic signal between FGFR2 WT and mutant cancers. All samples will undergo evaluation with RNASeq for gene expression. Results/Anticipated Results: Our initial analysis defined the range of RNA expression of FGFR2 and pFGFR2 protein signal (Y653/654 and Y769) between FGFR2 WT and FGFR2 mutant samples. On average, the FGFR2 mutant cohort displayed higher FGFR2 RNA expression compared to the FGFR2 WT cohort. There is no apparent correlation between RNA expression and clinical response to FGFR-targeted therapy. However, in this small cohort, there is no significant difference in FGFR2 phosphorylation between FGFR2 WT and mutant cancers. RPPA analysis of FGFR downstream signaling proteins reveals a wide range of phosphorylation, but no significant difference between FGFR2 WT and mutant cancers. Discussion/Significance of Impact: These findings illustrate the complexities of FGFR signaling between FGFR2 WT and mutant cancers. These data suggest that tumors with genomically WT FGFR may display increased pFGFR2 and downstream signaling phospho-proteins. We propose a larger study of cholangiocarcinoma to evaluate evidence of FGFR pathway activation in WT tumors.
Objectives/Goals: Unhealthy lifestyle habits may increase medical students’ risk for metabolic dysfunction-associated fatty liver disease (MAFLD) and non-alcoholic fatty pancreatic disease (NAFPD). This study aims to investigate how these lifestyle factors affect liver and pancreas health in preclinical medical students using diagnostic ultrasound imaging. Methods/Study Population: Using diagnostic ultrasound imaging, we propose a research study to evaluate the anatomical changes of the liver and pancreas associated with lifestyle among medical students in UCC. Forty-two (42) medical students from the Central University of the Caribbean who are in their preclinical years will be recruited to perform an abdominal ultrasound. To measure the diameter of the right liver lobe, we will employ the craniocaudal measurement method established by Riestra. et al. (2018). The parameter established by Rumack et al. (2011) will be utilized to assess liver texture and categories by Lee JS et al. (2009) to pancreas fat infiltration grades. Results/Anticipated Results: This study expects to reveal a significant correlation between the lifestyles of preclinical medical students and the health of their liver and pancreas, particularly in size and texture. We anticipate identifying specific lifestyle factors – such as dietary habits and physical activity levels – that contribute to the prevalence of hepatic and pancreatic steatosis. Additionally, we expect to highlight the need for targeted interventions to promote healthier lifestyles among medical students to mitigate risks associated with MAFLD and NAFPD. Discussion/Significance of Impact: This study is significant for monitoring changes in liver and pancreas health, preventing complications, and improving health quality while reducing future costs. It may guide the creation of tailored wellness programs for medical students, enhancing their well-being and contributing to better healthcare practices and educational strategies.
Objectives/Goals: This review evaluates recent advancements in artificial intelligence (AI)-driven radiomics for detecting early metabolic and structural changes via imaging techniques like PET-CT and magnetic resonance imaging (MRI). It aims to assess AI’s potential to predict disease progression and explore its implications for personalized preventative medicine. Methods/Study Population: This review analyzed studies from the past five years that explored AI applications in radiomics for early disease detection. The studies primarily focused on patients at risk for metabolic, cardiovascular, and oncological diseases. AI algorithms, including deep learning models, were evaluated for their ability to detect subtle metabolic and structural changes in imaging data from modalities like PET-CT and MRI. We categorized methodologies based on imaging biomarkers targeted, AI model architecture, and the clinical populations involved. The review highlights the methods used across studies to assess AI’s effectiveness in predicting disease progression. Results/Anticipated Results: The review found that AI models consistently demonstrated superior performance in detecting early metabolic and structural changes compared to traditional radiology methods. Across multiple studies, AI was able to identify biomarkers associated with disease progression months before clinical symptoms appeared, particularly in metabolic, cardiovascular, and cancer patients. Deep learning algorithms showed high accuracy in analyzing imaging data, improving predictive outcomes. The findings suggest that integrating AI into clinical practice could enable earlier interventions, offering personalized preventative care, and reducing the progression of late-stage diseases. Discussion/Significance of Impact: AI-driven radiomics holds great promise for transforming healthcare by enabling life-saving early detection and precision-based interventions. This technology could significantly reduce mortality in diseases like cancer, heart disease, and diabetes by allowing for earlier, targeted preventative strategies.
Objectives/Goals: Acute myeloid leukemia (AML) is the second most common leukemia among pediatric populations. Approximately 15% of pediatric AML cases have KMT2A gene rearrangements (KMT2A-r), which confers a worse prognosis. Our goal is to better characterize the biologic landscape of KMT2A-r pediatric AML. Methods/Study Population: This study utilizes deidentified peripheral blood and/or bone marrow samples banked in the Children’s Mercy Tumor Bank Biorepository. We investigated four KMT2A-r pediatric AML patients and six patients with other AML subtypes using samples collected at diagnosis and remission that were stored in the “tumor bank.” In addition, we assessed 47 tumor bank samples from patients with other leukemia subtypes. We performed differential expression (DE) analysis on bulk RNA sequencing comparing KMT2A-r and all other AML subtypes, as well as single-cell RNA sequencing and proteomic analysis on the larger cohort. We then coalesced these data to better identify processes and pathways that are dysregulated in KMT2A-r AML, specifically aiming to find those that were contributing to leukemogenesis. Results/Anticipated Results: Transcriptomic analysis showed that HOXA10 and MEIS1, two genes associated with immature myeloid populations and KMT2A-r leukemias, were more highly transcribed in AMLs than other leukemias. In addition, our DE analysis showed significantly higher transcription of ITGA7, a gene shown to correlate with poorer prognosis in AML, in our KMT2A-r samples when compared to other AML subtypes. FAM46C, a tumor suppressor gene contributing to mRNA stabilization, was less highly expressed in KMT2A-r AML when compared to other AML subtypes. Of note, low expression of FAM46C is associated with poorer survival and treatment response in multiple myeloma, and our findings suggest it may also be relevant to AML. Proteomic analysis is currently in process. Discussion/Significance of Impact: Transcriptomic analysis identifies unique molecular features of pediatric KMT2A-r AML. We anticipate that our proteomic data will do the same and will also corroborate our RNA findings. Taken in combination, these results will provide a more complete picture of the specific mechanisms contributing to this aggressive leukemic subtype.
Objectives/Goals: To develop a personalized computational framework integrating computational fluid dynamics (CFD) and topology optimization for designing intracranial aneurysm implants. The primary objective is to reduce intra-aneurysmal blood flow velocity and enhance thrombus formation for improved treatment outcomes. Methods/Study Population: Patient-specific aneurysm geometries were extracted from pre-treatment rotational angiograms. A CFD-driven topology optimization framework was employed to design implants that reduce intra-aneurysmal flow velocity. The fluid dynamics were modeled using Navier–Stokes equations and the structural integrity of the implants was ensured by linear elasticity equations. The solid isotropic material with penalization (SIMP) method was applied to optimize the implant’s porous architecture, balancing flow reduction with structural support. COMSOL Multiphysics software was used to implement the optimization. Results/Anticipated Results: The optimized implants demonstrated significant reductions in intra-aneurysmal blood flow velocity and improved hemodynamic conditions. Flow velocity within the aneurysm was reduced by 77%, and the fluid energy dissipation ratio showed a 78.9% improvement compared to pretreatment conditions. The optimized porous structures were tailored to the aneurysm’s specific geometry, providing personalized designs that improve flow stasis and thrombus formation. Further validation of the implants will be performed in vitro and in vivo to assess their effectiveness and biocompatibility. Discussion/Significance of Impact: This personalized implant design framework could lead to better treatment outcomes by reducing aneurysm recurrence and complications compared to current devices. It provides a pathway for improved occlusion rates and patient-specific solutions for intracranial aneurysms.
Objectives/Goals: Colorectal cancer (CRC) is classified into right-sided, left-sided, and rectal cancer. Clinicopathological and molecular features vary along the colorectum, even within subsites, leading to inconsistencies in identifying relevant biomarkers. We created a CRC metabolome map to explore diagnostic and survival heterogeneity across subsites. Methods/Study Population: A total of 372 patient-matched tumor and normal tissue samples were collected from seven colorectal subsites: cecum (n = 63), ascending colon (n = 44), transverse colon (n = 32), descending colon (n = 28), sigmoid colon (n = 75), rectosigmoid colon (n = 38), and rectum (n = 92). Liquid chromatography–mass spectrometry was used to compare metabolite abundances. Cox proportional hazards regression assessed metabolite impact on survival, adjusted for clinical covariates. Parametric and nonparametric tests were applied to compare the metabolite abundances. An interactive, publicly accessible online platform was developed to allow researchers to explore and generate hypotheses from this data. Results/Anticipated Results: Our study identified 39 and 70 significantly altered metabolites, including bile acids and lysophosphatidylcholines, across tumors and normal mucosa, showing metabolic heterogeneity between CRC subsites. We observed significant linear trends in metabolite gradients from the cecum to the rectum, and it was depended on the disease status. Comparison of tumors to patient-matched normal mucosa revealed metabolite changes exclusive to each subsite. Metabolite differences correlated with survival were unique to each subsite. Additionally, we developed an interactive, publicly accessible CRC metabolome database to share this valuable resource: https://colorectal-cancer-metabolome.com/yale-university. Discussion/Significance of Impact: This study provides the first CRC metabolome map, revealing metabolic differences across colorectal subsites. It challenges the right vs. left CRC classification, highlighting subsite-specific biomarker identification. Findings offer insights for personalized treatments tailored to the tumor type to improve patient outcomes.
Objectives/Goals: Lung transplant is a life-saving surgery for patients with advanced lung diseases yet long-term survival remains poor. The clinical features and lung injury patterns of lung transplant recipients who die early versus those who survive longer term remain undefined. Here, we use cell-free DNA and rejection parameters to help elucidate this further. Methods/Study Population: Lung transplant candidacy prioritizes patients who have a high mortality risk within 2 years and will likely survive beyond 5 years. We stratified patients who died within 2 years of transplant as early death (n = 50) and those who survived past 5 years as long-term survivors (n = 53). Lung transplant recipients had serial blood collected as part of two prospective cohort studies. Cell-free DNA (cfDNA) was quantified using relative (% donor-derived cfDNA {%ddcfDNA}) and absolute (nuclear-derived {n-cfDNA}, mitochondrial-derived {mt-cfDNA}) measurements. As part of routine posttransplant clinical care, all patients underwent pulmonary function testing (PFT), surveillance bronchoscopy with bronchoalveolar lavage (BAL), transbronchial biopsy (TBBx), and donor-specific antibody testing (DSA). Results/Anticipated Results: Over the first 2 years after transplant, the number of episodes of antibody-mediated rejection (p) Discussion/Significance of Impact: Clinically, early-death patients perform worse on routine surveillance PFTs and experience a worse degree of CLAD. These patients also have higher levels of cfDNA as quantified by n-cfDNA and mt-cfDNA. These results provide preliminary evidence that early-death patients have worse allograft rejection, dysfunction, and molecular injury.
Objectives/Goals: Identify risks in the regulatory reliance ecosystem, specifically for generic reliance pathways, using system theoretic process analysis (STPA), a proactive hazard analysis method, and create sustainable solutions to ensure global accessibility to the highest attainable standard of health. Methods/Study Population: A systematic literature review will assess the regulatory reliance ecosystem and interactions among National Regulatory Agencies (NRAs). This will involve using a hierarchical control structure (HCS) to identify operational loop failures, decision-making, and information flow. The HCS will inform an STPA to prevent adverse outcomes like patient harm or regulatory non-compliance. With IRB approval, 10 regulatory experts will also be interviewed to gather insights on risk scenarios. Their input will be used to integrate regulatory pathways into the HCS for adaptability, and a second round of interviews will be conducted to validate the scenarios and assess the effectiveness of recommendations. Results/Anticipated Results: A preliminary literature review on regulatory reliance pathways from PubMed, WHO guidelines, and the DIA global forum, revealed potential risks in NRA interactions, such as overly redacted reports obscuring critical details and discrepancies in product versions or incomplete reviews affecting evaluations. Diverging guidelines and failure to adapt to country-specific needs are potential risks that also impact patient access to essential medicines. The STPA framework, along with expert feedback, will uncover unknown risks like “secondary-reliance” and soft risks, leading to sustainable recommendations to improve and optimize the safety of regulatory reliance. Discussion/Significance of Impact: Safe regulatory reliance is crucial for global health equity and patient access to essential and innovative medicines. STPA will identify and address different layers of risk in the system, improving safety, efficiency, and innovation for timely patient access to therapeutics.