Try a new search

Format these results:

Searched for:

school:SOM

Department/Unit:Cell Biology

Total Results:

14036


Measuring the development of a medical professional identity through medical school

Lusk, P; Ark, T; Crowe, R; Monson, V; Altshuler, L; Harnik, V; Buckvar-Keltz, L; Poag, M; Belluomini, P; Kalet, A
PURPOSE/UNASSIGNED:The Professional Identity Essay (PIE) is a theory and evidence-based Medical Professional Identity Formation (MPIF) measure. We describe trajectories of PIE-measured MPIF over a 4-year US medical school curriculum. METHODS/UNASSIGNED:Students write PIEs at medical school orientation, clinical clerkships orientation, and post-advanced (near graduation) clerkship. A trained evaluator assigns an overall stage score to narrative responses to nine PIE prompts (inter-rater ICC 0.83, 95% CI [0.57 - 0.96], intra-rater ICC 0.85). Distribution of PIE stage scores across time points were analyzed in the aggregate and individual students were classified as Increase, Stable (no score change) or Decrease based on the trajectories of PIE stage scores over time. RESULTS/UNASSIGNED: CONCLUSIONS/UNASSIGNED:Medical students' PIE stage scores increase over time with three distinctive trajectories. Further study is needed to explore the utility of this method for formative assessment, program evaluation, and MPIF research.
PMID: 37917985
ISSN: 1466-187x
CID: 5655422

Integrative multi-omics profiling in human decedents receiving pig heart xenografts

Schmauch, Eloi; Piening, Brian; Mohebnasab, Maedeh; Xia, Bo; Zhu, Chenchen; Stern, Jeffrey; Zhang, Weimin; Dowdell, Alexa K; Kim, Jacqueline I; Andrijevic, David; Khalil, Karen; Jaffe, Ian S; Loza, Bao-Li; Gragert, Loren; Camellato, Brendan R; Oliveira, Michelli F; O'Brien, Darragh P; Chen, Han M; Weldon, Elaina; Gao, Hui; Gandla, Divya; Chang, Andrew; Bhatt, Riyana; Gao, Sarah; Lin, Xiangping; Reddy, Kriyana P; Kagermazova, Larisa; Habara, Alawi H; Widawsky, Sophie; Liang, Feng-Xia; Sall, Joseph; Loupy, Alexandre; Heguy, Adriana; Taylor, Sarah E B; Zhu, Yinan; Michael, Basil; Jiang, Lihua; Jian, Ruiqi; Chong, Anita S; Fairchild, Robert L; Linna-Kuosmanen, Suvi; Kaikkonen, Minna U; Tatapudi, Vasishta; Lorber, Marc; Ayares, David; Mangiola, Massimo; Narula, Navneet; Moazami, Nader; Pass, Harvey; Herati, Ramin S; Griesemer, Adam; Kellis, Manolis; Snyder, Michael P; Montgomery, Robert A; Boeke, Jef D; Keating, Brendan J
In a previous study, heart xenografts from 10-gene-edited pigs transplanted into two human decedents did not show evidence of acute-onset cellular- or antibody-mediated rejection. Here, to better understand the detailed molecular landscape following xenotransplantation, we carried out bulk and single-cell transcriptomics, lipidomics, proteomics and metabolomics on blood samples obtained from the transplanted decedents every 6 h, as well as histological and transcriptomic tissue profiling. We observed substantial early immune responses in peripheral blood mononuclear cells and xenograft tissue obtained from decedent 1 (male), associated with downstream T cell and natural killer cell activity. Longitudinal analyses indicated the presence of ischemia reperfusion injury, exacerbated by inadequate immunosuppression of T cells, consistent with previous findings of perioperative cardiac xenograft dysfunction in pig-to-nonhuman primate studies. Moreover, at 42 h after transplantation, substantial alterations in cellular metabolism and liver-damage pathways occurred, correlating with profound organ-wide physiological dysfunction. By contrast, relatively minor changes in RNA, protein, lipid and metabolism profiles were observed in decedent 2 (female) as compared to decedent 1. Overall, these multi-omics analyses delineate distinct responses to cardiac xenotransplantation in the two human decedents and reveal new insights into early molecular and immune responses after xenotransplantation. These findings may aid in the development of targeted therapeutic approaches to limit ischemia reperfusion injury-related phenotypes and improve outcomes.
PMID: 38760586
ISSN: 1546-170x
CID: 5654102

Sirt5 desuccinylates Cdc42 to mediate osteoclastogenesis and bone remodeling in mice

Zhang, Yuang; Wang, Jing; Luan, Jing; Liu, Chuanju; Cui, Yazhou; Han, Jinxiang
PMCID:10806281
PMID: 38274381
ISSN: 2352-3042
CID: 5625332

Impaired upper respiratory tract barrier function during postnatal development predisposes to invasive pneumococcal disease

Lokken-Toyli, Kristen L; Aggarwal, Surya D; Bee, Gavyn Chern Wei; de Steenhuijsen Piters, Wouter A A; Wu, Cindy; Chen, Kenny Zhi Ming; Loomis, Cynthia; Bogaert, Debby; Weiser, Jeffrey N
Infants are highly susceptible to invasive respiratory and gastrointestinal infections. To elucidate the age-dependent mechanism(s) that drive bacterial spread from the mucosa, we developed an infant mouse model using the prevalent pediatric respiratory pathogen, Streptococcus pneumoniae (Spn). Despite similar upper respiratory tract (URT) colonization levels, the survival rate of Spn-infected infant mice was significantly decreased compared to adults and corresponded with Spn dissemination to the bloodstream. An increased rate of pneumococcal bacteremia in early life beyond the newborn period was attributed to increased bacterial translocation across the URT barrier. Bacterial dissemination in infant mice was independent of URT monocyte or neutrophil infiltration, phagocyte-derived ROS or RNS, inflammation mediated by toll-like receptor 2 or interleukin 1 receptor signaling, or the pore-forming toxin pneumolysin. Using molecular barcoding of Spn, we found that only a minority of bacterial clones in the nasopharynx disseminated to the blood in infant mice, indicating the absence of robust URT barrier breakdown. Rather, transcriptional profiling of the URT epithelium revealed a failure of infant mice to upregulate genes involved in the tight junction pathway. Expression of many such genes was also decreased in early life in humans. Infant mice also showed increased URT barrier permeability and delayed mucociliary clearance during the first two weeks of life, which corresponded with tighter attachment of bacteria to the respiratory epithelium. Together, these results demonstrate a window of vulnerability during postnatal development when altered mucosal barrier function facilitates bacterial dissemination.
PMCID:11078396
PMID: 38718049
ISSN: 1553-7374
CID: 5658402

Autoimmune, Autoinflammatory Disease and Cutaneous Malignancy Associations with Hidradenitis Suppurativa: A Cross-Sectional Study

Brydges, Hilliard T; Onuh, Ogechukwu C; Friedman, Rebecca; Barrett, Joy; Betensky, Rebecca A; Lu, Catherine P; Caplan, Avrom S; Alavi, Afsaneh; Chiu, Ernest S
BACKGROUND:Hidradenitis suppurativa (HS) is a debilitating cutaneous disease characterized by severe painful inflammatory nodules/abscesses. At present, data regarding the epidemiology and pathophysiology of this disease are limited. OBJECTIVE:To define the prevalence and comorbidity associations of HS. METHODS:examining over 180 million US patients. Prevalences were calculated by demographic and odds ratios (OR) and identified comorbidity correlations. RESULTS:All examined metabolism-related, psychological, and autoimmune/autoinflammatory (AI) diseases correlated with HS. The strongest associations were with pyoderma gangrenosum [OR 26.56; confidence interval (CI): 24.98-28.23], Down syndrome (OR 11.31; CI 10.93-11.70), and polycystic ovarian syndrome (OR 11.24; CI 11.09-11.38). Novel AI associations were found between HS and lupus (OR 6.60; CI 6.26-6.94) and multiple sclerosis (MS; OR 2.38; CI 2.29-2.48). Cutaneous malignancies were largely not associated in the unsegmented cohort; however, among Black patients, novel associations with melanoma (OR 2.39; CI 1.86-3.08) and basal cell carcinoma (OR 2.69; CI 2.15-3.36) were identified. LIMITATIONS/CONCLUSIONS:International Classification of Diseases (ICD)-based disease identification relies on coding fidelity and diagnostic accuracy. CONCLUSION/CONCLUSIONS:This is the first study to identify correlations between HS with melanoma and basal cell carcinoma (BCC) among Black patients as well as MS and lupus in all patients with HS.
PMID: 38337127
ISSN: 1179-1888
CID: 5632132

Severity of COVID-19 Is Associated With Air Pollution: A Single Center Machine Learning Approach to Understand Risk

Kwon, S.; Zhao, Z.; Vora, K.; Crowley, G.; Podury, S.; Grunig, G.; Nolan, A.
ORIGINAL:0017189
ISSN: 2325-6621
CID: 5651802

Cytokine Receptor-like Factor 1 (CRLF1) and Its Role in Osteochondral Repair

Zhang, Fenglin; Clair, Andrew J; Dankert, John F; Lee, You Jin; Campbell, Kirk A; Kirsch, Thorsten
BACKGROUND:Since cytokine receptor-like factor 1 (CRLF1) has been implicated in tissue regeneration, we hypothesized that CRLF1 released by mesenchymal stem cells can promote the repair of osteochondral defects. METHODS:The degree of a femoral osteochondral defect repair in rabbits after intra-articular injections of bone marrow-derived mesenchymal stem cells (BMSCs) that were transduced with empty adeno-associated virus (AAV) or AAV containing CRLF1 was determined by morphological, histological, and micro computer tomography (CT) analyses. The effects of CRLF1 on chondrogenic differentiation of BMSCs or catabolic events of interleukin-1beta-treated chondrocyte cell line TC28a2 were determined by alcian blue staining, gene expression levels of cartilage and catabolic marker genes using real-time PCR analysis, and immunoblot analysis of Smad2/3 and STAT3 signaling. RESULTS:Intra-articular injections of BMSCs overexpressing CRLF1 markedly improved repair of a rabbit femoral osteochondral defect. Overexpression of CRLF1 in BMSCs resulted in the release of a homodimeric CRLF1 complex that stimulated chondrogenic differentiation of BMSCs via enhancing Smad2/3 signaling, whereas the suppression of CRLF1 expression inhibited chondrogenic differentiation. In addition, CRLF1 inhibited catabolic events in TC28a2 cells cultured in an inflammatory environment, while a heterodimeric complex of CRLF1 and cardiotrophin-like Cytokine (CLC) stimulated catabolic events via STAT3 activation. CONCLUSION/CONCLUSIONS:A homodimeric CRLF1 complex released by BMSCs enhanced the repair of osteochondral defects via the inhibition of catabolic events in chondrocytes and the stimulation of chondrogenic differentiation of precursor cells.
PMCID:11083199
PMID: 38727293
ISSN: 2073-4409
CID: 5656042

Introduction to the Compendium On Environmental Impacts on Cardiovascular Health and Biology [Editorial]

Hamburg, Naomi M; Moore, Kathryn J
PMID: 38662857
ISSN: 1524-4571
CID: 5657712

Conformational changes in the Niemann-Pick type C1 protein NCR1 drive sterol translocation

Frain, Kelly M; Dedic, Emil; Nel, Lynette; Bohush, Anastasiia; Olesen, Esben; Thaysen, Katja; Wüstner, Daniel; Stokes, David L; Pedersen, Bjørn Panyella
The membrane protein Niemann-Pick type C1 (NPC1, named NCR1 in yeast) is central to sterol homeostasis in eukaryotes. Saccharomyces cerevisiae NCR1 is localized to the vacuolar membrane, where it is suggested to carry sterols across the protective glycocalyx and deposit them into the vacuolar membrane. However, documentation of a vacuolar glycocalyx in fungi is lacking, and the mechanism for sterol translocation has remained unclear. Here, we provide evidence supporting the presence of a glycocalyx in isolated S. cerevisiae vacuoles and report four cryo-EM structures of NCR1 in two distinct conformations, named tense and relaxed. These two conformations illustrate the movement of sterols through a tunnel formed by the luminal domains, thus bypassing the barrier presented by the glycocalyx. Based on these structures and on comparison with other members of the Resistance-Nodulation-Division (RND) superfamily, we propose a transport model that links changes in the luminal domains with a cycle of protonation and deprotonation within the transmembrane region of the protein. Our model suggests that NPC proteins work by a generalized RND mechanism where the proton motive force drives conformational changes in the transmembrane domains that are allosterically coupled to luminal/extracellular domains to promote sterol transport.
PMCID:11009665
PMID: 38568972
ISSN: 1091-6490
CID: 5726232

The IRG1-itaconate axis protects from cholesterol-induced inflammation and atherosclerosis

Cyr, Yannick; Bozal, Fazli K; Barcia Durán, José Gabriel; Newman, Alexandra A C; Amadori, Letizia; Smyrnis, Panagiotis; Gourvest, Morgane; Das, Dayasagar; Gildea, Michael; Kaur, Ravneet; Zhang, Tracy; Wang, Kristin M; Von Itter, Richard; Schlegel, P Martin; Dupuis, Samantha D; Sanchez, Bernard F; Schmidt, Ann Marie; Fisher, Edward A; van Solingen, Coen; Giannarelli, Chiara; Moore, Kathryn J
Atherosclerosis is fueled by a failure to resolve lipid-driven inflammation within the vasculature that drives plaque formation. Therapeutic approaches to reverse atherosclerotic inflammation are needed to address the rising global burden of cardiovascular disease (CVD). Recently, metabolites have gained attention for their immunomodulatory properties, including itaconate, which is generated from the tricarboxylic acid-intermediate cis-aconitate by the enzyme Immune Responsive Gene 1 (IRG1/ACOD1). Here, we tested the therapeutic potential of the IRG1-itaconate axis for human atherosclerosis. Using single-cell RNA sequencing (scRNA-seq), we found that IRG1 is up-regulated in human coronary atherosclerotic lesions compared to patient-matched healthy vasculature, and in mouse models of atherosclerosis, where it is primarily expressed by plaque monocytes, macrophages, and neutrophils. Global or hematopoietic Irg1-deficiency in mice increases atherosclerosis burden, plaque macrophage and lipid content, and expression of the proatherosclerotic cytokine interleukin (IL)-1β. Mechanistically, absence of Irg1 increased macrophage lipid accumulation, and accelerated inflammation via increased neutrophil extracellular trap (NET) formation and NET-priming of the NLRP3-inflammasome in macrophages, resulting in increased IL-1β release. Conversely, supplementation of the Irg1-itaconate axis using 4-octyl itaconate (4-OI) beneficially remodeled advanced plaques and reduced lesional IL-1β levels in mice. To investigate the effects of 4-OI in humans, we leveraged an ex vivo systems-immunology approach for CVD drug discovery. Using CyTOF and scRNA-seq of peripheral blood mononuclear cells treated with plasma from CVD patients, we showed that 4-OI attenuates proinflammatory phospho-signaling and mediates anti-inflammatory rewiring of macrophage populations. Our data highlight the relevance of pursuing IRG1-itaconate axis supplementation as a therapeutic approach for atherosclerosis in humans.
PMCID:11009655
PMID: 38564634
ISSN: 1091-6490
CID: 5726212