EBioMedicinePub Date : 2024-11-09DOI: 10.1016/j.ebiom.2024.105438
Sebastian Einhauser, Claudia Asam, Manuela Weps, Antonia Senninger, David Peterhoff, Stilla Bauernfeind, Benedikt Asbach, George William Carnell, Jonathan Luke Heeney, Monika Wytopil, André Fuchs, Helmut Messmann, Martina Prelog, Johannes Liese, Samuel D Jeske, Ulrike Protzer, Michael Hoelscher, Christof Geldmacher, Klaus Überla, Philipp Steininger, Ralf Wagner
{"title":"Longitudinal effects of SARS-CoV-2 breakthrough infection on imprinting of neutralizing antibody responses.","authors":"Sebastian Einhauser, Claudia Asam, Manuela Weps, Antonia Senninger, David Peterhoff, Stilla Bauernfeind, Benedikt Asbach, George William Carnell, Jonathan Luke Heeney, Monika Wytopil, André Fuchs, Helmut Messmann, Martina Prelog, Johannes Liese, Samuel D Jeske, Ulrike Protzer, Michael Hoelscher, Christof Geldmacher, Klaus Überla, Philipp Steininger, Ralf Wagner","doi":"10.1016/j.ebiom.2024.105438","DOIUrl":"https://doi.org/10.1016/j.ebiom.2024.105438","url":null,"abstract":"<p><strong>Background: </strong>The impact of the infecting SARS-CoV-2 variant of concern (VOC) and the vaccination status was determined on the magnitude, breadth, and durability of the neutralizing antibody (nAb) profile in a longitudinal multicentre cohort study.</p><p><strong>Methods: </strong>173 vaccinated and 56 non-vaccinated individuals were enrolled after SARS-CoV-2 Alpha, Delta, or Omicron infection and visited four times within 6 months and nAbs were measured for D614G, Alpha, Delta, BA.1, BA.2, BA.5, BQ.1.1, XBB.1.5 and JN.1.</p><p><strong>Findings: </strong>Magnitude-breadth-analysis showed enhanced neutralization capacity in vaccinated individuals against multiple VOCs. Longitudinal analysis revealed sustained neutralization magnitude-breadth after antigenically distant Delta or Omicron breakthrough infection (BTI), with triple-vaccinated individuals showing significantly elevated titres and improved breadth. Antigenic mapping and antibody landscaping revealed initial boosting of vaccine-induced WT-specific responses after BTI, a shift in neutralization towards infecting VOCs at peak responses and an immune imprinted bias towards dominating WT immunity in the long-term. Despite that bias, machine-learning models confirmed a sustained shift of the immune-profiles following BTI.</p><p><strong>Interpretation: </strong>In summary, our longitudinal analysis revealed delayed and short lived nAb shifts towards the infecting VOC, but an immune imprinted bias towards long-term vaccine induced immunity after BTI.</p><p><strong>Funding: </strong>This work was funded by the Bavarian State Ministry of Science and the Arts for the CoVaKo study and the ForCovid project. The funders had no influence on the study design, data analysis or data interpretation.</p>","PeriodicalId":11494,"journal":{"name":"EBioMedicine","volume":"110 ","pages":"105438"},"PeriodicalIF":9.7,"publicationDate":"2024-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142616646","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
EBioMedicinePub Date : 2024-11-08DOI: 10.1016/j.ebiom.2024.105441
Tony Chen, Giang Pham, Louis Fox, Nina Adler, Xiaoyu Wang, Jingning Zhang, Jinyoung Byun, Younghun Han, Gretchen R B Saunders, Dajiang Liu, Michael J Bray, Alex T Ramsey, James McKay, Laura J Bierut, Christopher I Amos, Rayjean J Hung, Xihong Lin, Haoyu Zhang, Li-Shiun Chen
{"title":"Genomic insights for personalised care in lung cancer and smoking cessation: motivating at-risk individuals toward evidence-based health practices.","authors":"Tony Chen, Giang Pham, Louis Fox, Nina Adler, Xiaoyu Wang, Jingning Zhang, Jinyoung Byun, Younghun Han, Gretchen R B Saunders, Dajiang Liu, Michael J Bray, Alex T Ramsey, James McKay, Laura J Bierut, Christopher I Amos, Rayjean J Hung, Xihong Lin, Haoyu Zhang, Li-Shiun Chen","doi":"10.1016/j.ebiom.2024.105441","DOIUrl":"10.1016/j.ebiom.2024.105441","url":null,"abstract":"<p><strong>Background: </strong>Lung cancer and tobacco use pose significant global health challenges, necessitating a comprehensive translational roadmap for improved prevention strategies such as cancer screening and tobacco treatment, which are currently under-utilised. Polygenic risk scores (PRSs) may further motivate health behaviour change in primary care for lung cancer in diverse populations. In this work, we introduce the GREAT care paradigm, which integrates PRSs within comprehensive patient risk profiles to motivate positive health behaviour changes.</p><p><strong>Methods: </strong>We developed PRSs using large-scale multi-ancestry genome-wide association studies and standardised PRS distributions across all ancestries. We validated our PRSs in 561,776 individuals of diverse ancestry from the GISC Trial, UK Biobank (UKBB), and All of Us Research Program (AoU).</p><p><strong>Findings: </strong>Significant odds ratios (ORs) for lung cancer and difficulty quitting smoking were observed in both UKBB and AoU. For lung cancer, the ORs for individuals in the highest risk group (top 20% versus bottom 20%) were 1.85 (95% CI: 1.58-2.18) in UKBB and 2.39 (95% CI: 1.93-2.97) in AoU. For difficulty quitting smoking, the ORs (top 33% versus bottom 33%) were 1.36 (95% CI: 1.32-1.41) in UKBB and 1.32 (95% CI: 1.28-1.36) in AoU.</p><p><strong>Interpretation: </strong>Our PRS-based intervention model leverages large-scale genetic data for robust risk assessment across populations, which will be evaluated in two cluster-randomised clinical trials. This approach integrates genomic insights into primary care, promising improved outcomes in cancer prevention and tobacco treatment.</p><p><strong>Funding: </strong>National Institutes of Health, NIH Intramural Research Program, National Science Foundation.</p>","PeriodicalId":11494,"journal":{"name":"EBioMedicine","volume":"110 ","pages":"105441"},"PeriodicalIF":9.7,"publicationDate":"2024-11-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142616645","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
EBioMedicinePub Date : 2024-11-07DOI: 10.1016/j.ebiom.2024.105431
Hasan Farooq, Hannah P Luehmann, Jeffrey R Koenitzer, Gyu Seong Heo, Deborah H Sultan, Devesha H Kulkarni, Sean P Gunsten, Rekha M Sashti, Tao Huang, Amanda R Keller, Kory J Lavine, Jeffrey J Atkinson, Laura M Wingler, Yongjian Liu, Steven L Brody
{"title":"Molecular imaging in experimental pulmonary fibrosis reveals that nintedanib unexpectedly modulates CCR2 immune cell infiltration.","authors":"Hasan Farooq, Hannah P Luehmann, Jeffrey R Koenitzer, Gyu Seong Heo, Deborah H Sultan, Devesha H Kulkarni, Sean P Gunsten, Rekha M Sashti, Tao Huang, Amanda R Keller, Kory J Lavine, Jeffrey J Atkinson, Laura M Wingler, Yongjian Liu, Steven L Brody","doi":"10.1016/j.ebiom.2024.105431","DOIUrl":"https://doi.org/10.1016/j.ebiom.2024.105431","url":null,"abstract":"<p><strong>Background: </strong>Pulmonary fibrosis is a challenging clinical problem with lung pathology featuring immune cell infiltrates, fibroblast expansion, and matrix deposition. Molecular analysis of diseased lungs and preclinical models have uncovered C-C chemokine receptor type 2 (CCR2)+ monocyte egress from the bone marrow into the lung, where they acquire profibrotic activities. Current drug treatment is focused on fibroblast activity. Alternatively, therapeutic targeting and monitoring CCR2+ cells may be an effective patient management strategy.</p><p><strong>Methods: </strong>Inhibition of CCR2+ cells and, as a benchmark, the clinical antifibrotic agent, nintedanib, were used in mouse lung fibrosis models. Lungs were evaluated directly for CCR2+ cell infiltration and by non-invasive CCR2+ positron emission tomography imaging (CCR2-PET).</p><p><strong>Findings: </strong>Lung CCR2+ cells were significantly elevated in the bleomycin model as determined by tissue evaluation and CCR2-PET imaging. A protective treatment protocol with an oral CCR2 inhibitor was compared to oral nintedanib. While we expected disparate effects on CCR2+ cells, each drug similarly decreased lung CCR2+ cells and fibrosis. Chemotaxis assays showed nintedanib indirectly inhibited C-C motif chemokine 2 (CCL2)-mediated migration of CCR2+ cells. Even delayed therapeutic administration of nintedanib in bleomycin and the silicosis progressive fibrosis models decreased the accumulation of CCR2+ lung cells. In these treatments early CCR2-PET imaging predicted the later development of fibrosis.</p><p><strong>Interpretation: </strong>The inhibition of CCR2+ cell egress is likely a critical controller for stabilising lung fibrosis, as provided by nintedanib. Imaging with CCR2-PET may be useful to monitor nintedanib treatment responses, guide decision-making in the treatment of patients with progressive pulmonary fibrosis, and as a biomarker for drug development.</p><p><strong>Funding: </strong>National Institutes of Health (NIH), R01HL131908 (SLB), R35HL145212 (YL), P41EB025815 (YL), K01DK133670 (DHK); Barnes Jewish Hospital Foundation (SLB).</p>","PeriodicalId":11494,"journal":{"name":"EBioMedicine","volume":"110 ","pages":"105431"},"PeriodicalIF":9.7,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142603846","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
EBioMedicinePub Date : 2024-11-07DOI: 10.1016/j.ebiom.2024.105434
Ingrid Berg, Pia Härvelid, Wolfgang Emanuel Zürrer, Marianna Rosso, Daniel S Reich, Benjamin Victor Ineichen
{"title":"Which experimental factors govern successful animal-to-human translation in multiple sclerosis drug development? A systematic review and meta-analysis.","authors":"Ingrid Berg, Pia Härvelid, Wolfgang Emanuel Zürrer, Marianna Rosso, Daniel S Reich, Benjamin Victor Ineichen","doi":"10.1016/j.ebiom.2024.105434","DOIUrl":"https://doi.org/10.1016/j.ebiom.2024.105434","url":null,"abstract":"<p><strong>Background: </strong>Despite successes in multiple sclerosis (MS) drug development, the effectiveness of animal studies in predicting successful bench-to-bedside translation is uncertain. Our goal was to identify predictors of successful animal-to-human translation for MS by systematically comparing animal studies of approved disease-modifying therapies (DMTs) with those that failed in clinical trials due to efficacy or safety concerns.</p><p><strong>Methods: </strong>Systematic review of animal studies testing MS DMTs, identified from searches in PubMed and EMBASE. A random effect meta-analysis was fitted to the data to compare outcome effect sizes for approved versus failed DMTs. Effect sizes and testing under diverse experimental conditions were assessed as potential predictors for successful translation.</p><p><strong>Findings: </strong>We included 497 animal studies, covering 15 approved and 11 failed DMTs, tested in approximately 30'000 animals. DMTs were tested in a small repertoire of experimental parameters: about 86% of studies used experimental autoimmune encephalomyelitis (EAE), 80% used mice, and 76% used female animals. There was no association between animal study outcomes or testing DMTs under varied conditions (e.g., different laboratories or models) and successful approval. Surprisingly, 91% of animal studies were published after first-in-MS trial and 91% after official regulatory approval.</p><p><strong>Interpretation: </strong>Our findings emphasize the complexity in carrying drugs from animals to clinical practice. Specific challenges include limited experimental methods in animal research and a disconnect between preclinical and clinical research. We advocate for efforts to streamline drug development for MS to improve animal research's relevance for patients.</p><p><strong>Funding: </strong>NIH, Swiss National Science Foundation, Universities Federation for Animal Welfare.</p>","PeriodicalId":11494,"journal":{"name":"EBioMedicine","volume":"110 ","pages":"105434"},"PeriodicalIF":9.7,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142603849","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
EBioMedicinePub Date : 2024-11-07DOI: 10.1016/j.ebiom.2024.105444
Emma R Sutton, Ariane Beauvais, Rebecca Yaworski, Yves De Repentigny, Aoife Reilly, Monique Marylin Alves de Almeida, Marc-Olivier Deguise, Kathy L Poulin, Robin J Parks, Bernard L Schneider, Rashmi Kothary
{"title":"Liver SMN restoration rescues the Smn<sup>2B/-</sup> mouse model of spinal muscular atrophy.","authors":"Emma R Sutton, Ariane Beauvais, Rebecca Yaworski, Yves De Repentigny, Aoife Reilly, Monique Marylin Alves de Almeida, Marc-Olivier Deguise, Kathy L Poulin, Robin J Parks, Bernard L Schneider, Rashmi Kothary","doi":"10.1016/j.ebiom.2024.105444","DOIUrl":"https://doi.org/10.1016/j.ebiom.2024.105444","url":null,"abstract":"<p><strong>Background: </strong>The liver is a key metabolic organ, acting as a hub to metabolically connect various tissues. Spinal muscular atrophy (SMA) is a neuromuscular disorder whereby patients have an increased susceptibility to developing dyslipidaemia and liver steatosis. It remains unknown whether fatty liver is due to an intrinsic or extrinsic impact of survival motor neuron (SMN) protein depletion.</p><p><strong>Methods: </strong>Using an adeno-associated viral vector with a liver specific promoter (albumin), we restored SMN protein levels in the liver alone in Smn<sup>2B/-</sup> mice, a model of SMA. Experiments assessed central and peripheral impacts using immunoblot, immunohistochemistry, and electron microscopy techniques.</p><p><strong>Findings: </strong>We demonstrate that AAV9-albumin-SMN successfully expresses SMN protein in the liver with no detectable expression in the spinal cord or muscle in Smn<sup>2B/-</sup> mice. Liver intrinsic rescue of SMN protein was sufficient to increase survival of Smn<sup>2B/-</sup> mice. Fatty liver was ameliorated while key markers of liver function were also restored to normal levels. Certain peripheral pathologies were rescued including muscle size and pancreatic cell imbalance. Only a partial CNS recovery was seen using a liver therapeutic strategy alone.</p><p><strong>Interpretation: </strong>The fatty liver phenotype is a direct impact of liver intrinsic SMN protein loss. Correction of SMN protein levels in liver is enough to restore some aspects of disease in SMA. We conclude that the liver is an important contributor to whole-body pathology in Smn<sup>2B/-</sup> mice.</p><p><strong>Funding: </strong>This work was funded by Muscular Dystrophy Association (USA) [grant number 963652 to R.K.]; the Canadian Institutes of Health Research [grant number PJT-186300 to R.K.].</p>","PeriodicalId":11494,"journal":{"name":"EBioMedicine","volume":"110 ","pages":"105444"},"PeriodicalIF":9.7,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142603845","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
EBioMedicinePub Date : 2024-11-06DOI: 10.1016/j.ebiom.2024.105429
Matthew Moll, Julian Hecker, John Platig, Jingzhou Zhang, Auyon J Ghosh, Katherine A Pratte, Rui-Sheng Wang, Davin Hill, Iain R Konigsberg, Joe W Chiles, Craig P Hersh, Peter J Castaldi, Kimberly Glass, Jennifer G Dy, Don D Sin, Ruth Tal-Singer, Majd Mouded, Stephen I Rennard, Gary P Anderson, Gregory L Kinney, Russell P Bowler, Jeffrey L Curtis, Merry-Lynn McDonald, Edwin K Silverman, Brian D Hobbs, Michael H Cho
{"title":"Polygenic and transcriptional risk scores identify chronic obstructive pulmonary disease subtypes in the COPDGene and ECLIPSE cohort studies.","authors":"Matthew Moll, Julian Hecker, John Platig, Jingzhou Zhang, Auyon J Ghosh, Katherine A Pratte, Rui-Sheng Wang, Davin Hill, Iain R Konigsberg, Joe W Chiles, Craig P Hersh, Peter J Castaldi, Kimberly Glass, Jennifer G Dy, Don D Sin, Ruth Tal-Singer, Majd Mouded, Stephen I Rennard, Gary P Anderson, Gregory L Kinney, Russell P Bowler, Jeffrey L Curtis, Merry-Lynn McDonald, Edwin K Silverman, Brian D Hobbs, Michael H Cho","doi":"10.1016/j.ebiom.2024.105429","DOIUrl":"10.1016/j.ebiom.2024.105429","url":null,"abstract":"<p><strong>Background: </strong>Genetic variants and gene expression predict risk of chronic obstructive pulmonary disease (COPD), but their effect on COPD heterogeneity is unclear. We aimed to define high-risk COPD subtypes using genetics (polygenic risk score, PRS) and blood gene expression (transcriptional risk score, TRS) and assess differences in clinical and molecular characteristics.</p><p><strong>Methods: </strong>We defined high-risk groups based on PRS and TRS quantiles by maximising differences in protein biomarkers in a COPDGene training set and identified these groups in COPDGene and ECLIPSE test sets. We tested multivariable associations of subgroups with clinical outcomes and compared protein-protein interaction networks and drug repurposing analyses between high-risk groups.</p><p><strong>Findings: </strong>We examined two high-risk omics-defined groups in non-overlapping test sets (n = 1133 NHW COPDGene, n = 299 African American (AA) COPDGene, n = 468 ECLIPSE). We defined \"high activity\" (low PRS, high TRS) and \"severe risk\" (high PRS, high TRS) subgroups. Participants in both subgroups had lower body-mass index (BMI), lower lung function, and alterations in metabolic, growth, and immune signalling processes compared to a low-risk (low PRS, low TRS) subgroup. \"High activity\" but not \"severe risk\" participants had greater prospective FEV<sub>1</sub> decline (COPDGene: -51 mL/year; ECLIPSE: -40 mL/year) and proteomic profiles were enriched in gene sets perturbed by treatment with 5-lipoxygenase inhibitors and angiotensin-converting enzyme (ACE) inhibitors.</p><p><strong>Interpretation: </strong>Concomitant use of polygenic and transcriptional risk scores identified clinical and molecular heterogeneity amongst high-risk individuals. Proteomic and drug repurposing analysis identified subtype-specific enrichment for therapies and suggest prior drug repurposing failures may be explained by patient selection.</p><p><strong>Funding: </strong>National Institutes of Health.</p>","PeriodicalId":11494,"journal":{"name":"EBioMedicine","volume":"110 ","pages":"105429"},"PeriodicalIF":9.7,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11570824/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142603847","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Identifying WHO global priority endemic pathogens for vaccine research and development (R&D) using multi-criteria decision analysis (MCDA): an objective of the Immunization Agenda 2030.","authors":"Mateusz Hasso-Agopsowicz, Angela Hwang, Maria-Graciela Hollm-Delgado, Isis Umbelino-Walker, Ruth A Karron, Raman Rao, Kwaku Poku Asante, Meru Sheel, Erin Sparrow, Birgitte Giersing","doi":"10.1016/j.ebiom.2024.105424","DOIUrl":"https://doi.org/10.1016/j.ebiom.2024.105424","url":null,"abstract":"<p><strong>Background: </strong>To date, global priorities for new vaccine R&D have not been systematically identified for endemic pathogens. As part of Immunisation Agenda 2030 (IA2030), we have systematically identified priority endemic pathogens for new vaccine R&D based on country and regional stakeholder values to address this need.</p><p><strong>Methods: </strong>MCDA surveys targeting policy makers and immunisation stakeholders in each World Health Organization (WHO) region were used to weight eight criteria for prioritisation. Applying those weights to regional pathogen data yielded regional top ten pathogen lists, which are intended to inform regional deliberations on R&D priorities. The regional top ten lists were combined into an IA2030 global priority list. To inform R&D, use cases for new vaccines and monoclonal antibodies were identified, then categorized in terms of the activities needed to accelerate progress.</p><p><strong>Findings: </strong>In five out of six WHO regions, Annual deaths in children under five and Contribution to antimicrobial resistance were the most heavily weighted criteria. How participants weighted the criteria was not associated with their region, biographical characteristics, or areas of expertise. Five pathogens were common priorities across all regions: M tuberculosis, HIV-1, K pneumoniae, S aureus, and Extra-intestinal pathogenic E coli. Six pathogens were priorities in single regions. Combining regional top ten lists provided a global list of 17 priority pathogens for new vaccine R&D. Thirty-four distinct use cases were identified for new products targeting these pathogens. While most are in the \"Advance product development\" category, ten are in the \"Research\" category and seven are in the \"Prepare to implement\" category.</p><p><strong>Interpretation: </strong>These priorities for new vaccine R&D will help stakeholders better respond to regional and country needs. The use cases will inform R&D and enable monitoring of R&D under IA2030.</p><p><strong>Funding: </strong>The work was funded by a Bill and Melinda Gates Foundation grant to WHO (INV-005318).</p>","PeriodicalId":11494,"journal":{"name":"EBioMedicine","volume":" ","pages":"105424"},"PeriodicalIF":9.7,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142582338","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
EBioMedicinePub Date : 2024-11-04DOI: 10.1016/j.ebiom.2024.105433
Laura Xicota, Lam-Ha T Dang, Alice Lee, Sharon Krinsky-McHale, Deborah Pang, Lisa Melilli, Sid O'Bryant, Rachel L Henson, Charles Laymon, Florence Lai, H Diana Rosas, Beau Ances, Ira Lott, Christy Hom, Bradley Christian, Sigan Hartley, Shahid Zaman, Elizabeth Head, Mark Mapstone, Zhezhen Jin, Wayne Silverman, Nicole Schupf, Benjamin Handen, Joseph H Lee
{"title":"The effects of mosaicism on biological and clinical markers of Alzheimer's disease in adults with Down syndrome.","authors":"Laura Xicota, Lam-Ha T Dang, Alice Lee, Sharon Krinsky-McHale, Deborah Pang, Lisa Melilli, Sid O'Bryant, Rachel L Henson, Charles Laymon, Florence Lai, H Diana Rosas, Beau Ances, Ira Lott, Christy Hom, Bradley Christian, Sigan Hartley, Shahid Zaman, Elizabeth Head, Mark Mapstone, Zhezhen Jin, Wayne Silverman, Nicole Schupf, Benjamin Handen, Joseph H Lee","doi":"10.1016/j.ebiom.2024.105433","DOIUrl":"10.1016/j.ebiom.2024.105433","url":null,"abstract":"<p><strong>Background: </strong>Individuals with Down syndrome (DS) are at high risk of early-onset Alzheimer's disease (AD); yet, some 20 percent do not develop any signs of dementia until after 65 years or in their lifetime. Mosaicism could contribute to this phenotypic variation, where some disomic cells could lead to lower levels of gene products from chromosome 21.</p><p><strong>Methods: </strong>We examined longitudinal neuropsychological and biomarker data from two large studies of DS: the Alzheimer Biomarker Consortium-Down syndrome study (ABC-DS) (n = 357); and a legacy study (n = 468). We assessed mosaicism using karyotyping or GWAS data. Participants had data on plasma AD biomarkers (Aβ<sub>40</sub>, Aβ<sub>42</sub>, tau, and NfL) and longitudinal cognitive measures. A subset had cerebrospinal fluid biomarkers (Aβ<sub>40</sub>, Aβ<sub>42</sub>, tau, ptau181, and NfL) and amyloid and tau PET data.</p><p><strong>Findings: </strong>For both cohorts, the prevalence of mosaicism was <10% (ABC-DS: 7.3%; Legacy: 9.6%), and those with mosaicism had lower plasma Aβ<sub>40</sub> and Aβ<sub>42</sub> concentrations. For the older legacy cohort, when compared to those with full trisomy, those with mosaicism had significantly smaller decline in total and annualized neurocognitive scores, and lower incidence and prevalence of dementia.</p><p><strong>Interpretation: </strong>Mosaicism in DS was associated with lower concentrations of plasma Aβ peptides, possibly leading to lower AD risk. However, its clinical impact was less clear in the younger ABC-DC cohort, and a follow-up study is warranted.</p><p><strong>Funding: </strong>National Institutes of Health (R01AG014673, P01HD035897, R56AG061837), NIA (U01AG051412, U19AG068054), NICHD, ADRC programs, the Eunice Kennedy Shriver Intellectual and Developmental Disabilities Research Centers Program, and NCATS (UL1TR001873).</p>","PeriodicalId":11494,"journal":{"name":"EBioMedicine","volume":"110 ","pages":"105433"},"PeriodicalIF":9.7,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11570228/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142582456","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
EBioMedicinePub Date : 2024-11-01Epub Date: 2024-10-03DOI: 10.1016/j.ebiom.2024.105382
Katrin D Mayer-Barber, Daniel L Barber, Laura E Via
{"title":"Exhausted CD8 T cells and anti-inflammatory macrophages characterize lepromatous leprosy lesions.","authors":"Katrin D Mayer-Barber, Daniel L Barber, Laura E Via","doi":"10.1016/j.ebiom.2024.105382","DOIUrl":"10.1016/j.ebiom.2024.105382","url":null,"abstract":"","PeriodicalId":11494,"journal":{"name":"EBioMedicine","volume":"109 ","pages":"105382"},"PeriodicalIF":9.7,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11489073/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142375261","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Integrated single-cell transcriptomic analyses identify a novel lineage plasticity-related cancer cell type involved in prostate cancer progression.","authors":"Faming Zhao, Tingting Zhang, Jinlan Wei, Liang Chen, Zaoqu Liu, Yang Jin, Mingsheng Liu, Hongqing Zhou, Yanxia Hu, Xia Sheng","doi":"10.1016/j.ebiom.2024.105398","DOIUrl":"10.1016/j.ebiom.2024.105398","url":null,"abstract":"<p><strong>Background: </strong>Cancer cell plasticity is the ability of neoplastic cells to alter their identity and acquire new biological properties under microenvironmental pressures. In prostate cancer (PCa), lineage plasticity often results in therapy resistance and trans-differentiation to neuroendocrine (NE) lineage. However, identifying the cancer cells harboring lineage plasticity-related status remains challenging.</p><p><strong>Methods: </strong>Based on 13 multi-center human PCa bulk transcriptomic cohorts (samples = 3314) and 9 bulk transcriptomic datasets derived from PCa experimental models, we established an integrated lineage plasticity-related gene signature, termed LPSig. Leveraging this gene signature, AUCell enrichment analysis was applied to identify the cell population with high lineage plasticity from a comprehensive single-cell RNA-sequencing (scRNA-seq) meta-atlas assembled by us, which consisted of 10 public human PCa scRNA-seq datasets (samples = 93, cells = 222,529). Moreover, additional scRNA-seq dataset of human PCa, multiplex immunohistochemistry staining for human PCa tissues, in vitro and in vivo functional experiments, as well as qPCR and Western blot analyses were employed to validate our findings.</p><p><strong>Findings: </strong>We found that LPSig could finely capture the dynamics of tumor lineage plasticity throughout the progression of PCa, accurately estimating the status of lineage plasticity. Based on LPSig, we identified a previously undefined minority population of lineage plasticity-related PCa cells (LPCs) from the human PCa scRNA-seq meta-atlas assembled by this study. Furthermore, in-depth dissection revealed pivotal roles of LPCs in trans-differentiation, tumor recurrence, and poor patient survival during PCa progression. Furthermore, we identified HMMR as a representative cell surface marker for LPCs, which was validated using additional scRNA-seq datasets and multiplexed immunohistochemistry. Moreover, HMMR was transcriptionally inhibited by androgen receptor (AR), and was required for the aggressive adenocarcinoma features and NE phenotype.</p><p><strong>Interpretation: </strong>Our study uncovers a novel population of lineage plasticity-related cells with low AR activity, stemness-like traits, and elevated HMMR expression, that may facilitate poor prognosis in PCa.</p><p><strong>Funding: </strong>This work was supported by National Key R&D Program of China (2022YFA0807000), National Natural Science Foundation of China (82160584), Advanced Prostate Cancer Diagnosis and Treatment Technology Innovation Team of Kunming Medical University (CXTD202216), and Reserve Talents of Young and Middle-aged Academic Leaders in Yunnan Province (202105AC160013).</p>","PeriodicalId":11494,"journal":{"name":"EBioMedicine","volume":"109 ","pages":"105398"},"PeriodicalIF":9.7,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11530610/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142460704","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}