Rebeca Linhares Abreu Netto,Catherine Chen,Victor Irungu Mwangi,Carlos Eduardo Padron de Morais,Mariana Simão Xavier,Luiz Gustavo Gardinassi,Emily Marie Eriksson,Nicholas Kiernan-Walker,Eamon Conway,Ivo Mueller,Marcus Vinícius Guimarães Lacerda,Wuelton Marcelo Monteiro,Fernando Fonseca Almeida-Val,Gisely Cardoso de Melo
{"title":"Autoantibodies against type I interferons are a prominent feature in SARS-CoV-2 fatal disease and hospitalization.","authors":"Rebeca Linhares Abreu Netto,Catherine Chen,Victor Irungu Mwangi,Carlos Eduardo Padron de Morais,Mariana Simão Xavier,Luiz Gustavo Gardinassi,Emily Marie Eriksson,Nicholas Kiernan-Walker,Eamon Conway,Ivo Mueller,Marcus Vinícius Guimarães Lacerda,Wuelton Marcelo Monteiro,Fernando Fonseca Almeida-Val,Gisely Cardoso de Melo","doi":"10.1093/infdis/jiaf514","DOIUrl":"https://doi.org/10.1093/infdis/jiaf514","url":null,"abstract":"BACKGROUNDAutoantibodies have been implicated as key players in COVID-19 pathogenesis, with evidence of impacting disease severity and poor outcomes.METHODSWe analyzed autoantibody profiles in 435 PCR-confirmed COVID-19 patients in Manaus, Brazil, comparing hospitalized (263) and non-hospitalized (172) individuals, and 37 healthy controls. Twenty autoantibodies and SARS-CoV-2-specific IgA/IgG antibodies were measured using multiplex Luminex® assays from plasma samples collected on days 1, 7, 14, and 28.RESULTSHospitalized patients had significantly higher levels of autoantibodies targeting ACE2, MPO, IFNΩ, IFNα1 and IFNα2 at baseline, with 127 hospitalized patients positive for IFNα1, 161 for IFNα2 and 68 for IFNΩ. Longitudinal analysis revealed progressively increasing levels of anti-ACE2, B2G1, C1q, IFNα1, IFNα2, PADI4, PF4 and PR3 autoantibodies in hospitalized patients, however the neutralizing capacity of IFNs were not investigated. Survival analysis showed that elevated type I IFN autoantibodies correlated with reduced survival (p=0.023). Stronger correlations between autoantibodies and SARS-CoV-2 IgA/IgG were found in severe cases.CONCLUSIONSIn this cohort, autoantibodies were linked to COVID-19 severity and mortality, indicating their potential as biomarkers for patient risk stratification and therapeutic targets. Further research is needed to explore their impact in long-term immune dysregulation and post-acute sequelae in COVID-19 survivors.","PeriodicalId":501010,"journal":{"name":"The Journal of Infectious Diseases","volume":"53 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-10-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145261509","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Single-cell transcriptome profiling reveals the immune dysregulation characteristics of mice infected by Brucella abortus.","authors":"Guangzhi Zhang,Qingchun Shen,Jianxin Ye,Yu Feng,Pascal Boireau,Xuezheng Fan,Lang Lv,Yan Li,Xiaofeng Xu,Heleer Cha,Chenguang Shen,Yinghui Zhang,Xiaowei Peng,Hui Jiang,Jiabo Ding","doi":"10.1093/infdis/jiaf522","DOIUrl":"https://doi.org/10.1093/infdis/jiaf522","url":null,"abstract":"BACKGROUNDBrucellosis poses a significant threat to animal and human health globally. However, how Brucella subverts the immune response to establish persistent infections remains unclear.METHODSWe utilized single-cell RNA sequencing (scRNA-seq) to decipher the immune landscape of mice infected by Brucella abortus. Flow cytometry, transgenic cell line and mouse, and antibody blockage were utilized to explore the relevant mechanisms.RESULTSBrucella infection induced significant changes in the composition and signaling pathways of immune cells, and flow cytometry analysis further confirmed the scRNA-seq data. An in-depth analysis with macrophages, the main target cell for Brucella, demonstrated activation of type I IFN and type II IFN signaling, TNF production, diverse cell deaths, etc. Specifically, Vir-2308 Brucella infection induced IFN-β expression, primarily originating from macrophages. In vitro, a significantly lower level of intracellular Brucella survival was observed in ifnar1-/- macrophages. In vivo, ifnar1 genetic deficiency rendered the mice less susceptible to Brucella challenge with a lower bacterial load and higher levels of macrophages and neutrophils. Interestingly, Brucella infection induced a dramatic reduction of NK cells along with the upregulation of CD94:NKG2A, one typical immune checkpoint module of NK cells. Further blockage of the NKG2A receptor in mice significantly reduced the bacterial load in the tissues, concurrent with a higher ratio of mature dendritic cells and a lower proportion of B cells.CONCLUSIONSscRNA-seq revealed that Brucella infection significantly alters the immune microenvironment in mice, providing insight into a better understanding of brucellosis pathogenesis and the immune evasion strategies of this sophisticated pathogen.","PeriodicalId":501010,"journal":{"name":"The Journal of Infectious Diseases","volume":"72 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-10-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145261508","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Edward P Acosta, Inmaculada Aban, Kevin J Ryan, Pablo J Sánchez, Carina A Rodriguez, José R Romero, Sunil K Sood, Nazha Abughali, Ravit Arav-Boger, Roberta L DeBiasi, Gregory A Storch, John A Vanchiere, Kalyani Peri, Richard J Whitley, David W Kimberlin
{"title":"Ganciclovir Dosing in Premature Infants Receiving Treatment for Congenital Cytomegalovirus Infection: Results of a Prospective Pharmacokinetic Study","authors":"Edward P Acosta, Inmaculada Aban, Kevin J Ryan, Pablo J Sánchez, Carina A Rodriguez, José R Romero, Sunil K Sood, Nazha Abughali, Ravit Arav-Boger, Roberta L DeBiasi, Gregory A Storch, John A Vanchiere, Kalyani Peri, Richard J Whitley, David W Kimberlin","doi":"10.1093/infdis/jiaf528","DOIUrl":"https://doi.org/10.1093/infdis/jiaf528","url":null,"abstract":"Background Ganciclovir remains the primary therapeutic for cytomegalovirus (CMV) infections in early infancy, but its pharmacokinetics and dosing in very preterm infants with end-organ CMV disease have not been fully evaluated. Methods Premature infants with confirmed CMV infection and receiving ganciclovir as standard of care were enrolled into a pharmacokinetic sampling study. All were <32 weeks gestational age and >500 grams at enrollment. Plasma for ganciclovir quantitation was collected at steady-state at 0, 1, and between 2-3, 5-7, and 10-12 hours post-dose. Specimens were shipped, analyzed, and pharmacokinetic parameters calculated in real-time. Noncompartmental and modeling approaches were used for analysis. Results Eighteen infants were enrolled; mean gestational age at delivery was 26.7 weeks, and mean age and weight at enrollment were 42 days and 1519.0 grams, respectively. Seventeen completed pharmacokinetic assessments. Geometric mean dose and resulting 12 hour area-under-the-curve (AUC12) were 5.19 mg/kg and 52.7 mgxh/L, respectively. A total of 85 ganciclovir concentration-time data points were available for modeling. A one-compartment power covariate model with weight and serum creatinine on clearance and weight on distribution volume was used. Noncompartmental and modeled pharmacokinetic parameters were similar. Conclusions These are the first intravenous ganciclovir population pharmacokinetic data with covariate assessments in premature infants being treated for CMV disease. Results suggest an intravenous dose of 5 mg/kg/dose every 12 hours may be an appropriate starting regimen for treatment of premature infants with congenital CMV. Additional data are needed in this and other populations to better define optimal ganciclovir exposure targets.","PeriodicalId":501010,"journal":{"name":"The Journal of Infectious Diseases","volume":"10 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145260724","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Kevin D He, Simon B Gressens, Darshana M Dadhania, Hannah M Gilligan, Caitlin Davis, Emmanuel Edusei, Jenny Ahn, Michelle Lifton, Diana V Pastrana, Steven Kleiboeker, Christopher B Buck, Stephanie Jost, David Wojciechowski, C Sabrina Tan
{"title":"Immune responses to BK virus in renal transplant recipients receiving IVIG treatment","authors":"Kevin D He, Simon B Gressens, Darshana M Dadhania, Hannah M Gilligan, Caitlin Davis, Emmanuel Edusei, Jenny Ahn, Michelle Lifton, Diana V Pastrana, Steven Kleiboeker, Christopher B Buck, Stephanie Jost, David Wojciechowski, C Sabrina Tan","doi":"10.1093/infdis/jiaf525","DOIUrl":"https://doi.org/10.1093/infdis/jiaf525","url":null,"abstract":"Background BK polyomavirus (BKPyV) DNAemia in renal transplant recipients increases risk of allograft failure, but BKPyV-specific therapies are not available. While treatment with intravenous immune globulin (IVIG) has been reported, safety in a prospective randomized controlled setting is uncertain, and host immune response to BKV after IVIG is not well characterized. Methods We investigated host immune responses to BKPyV in a multicenter, prospective, randomized, double-blinded, placebo-controlled pilot study of IVIG with protocolized immunosuppression reduction in adult kidney transplant recipients with BK DNAemia. Participants received two infusions of 1g/kg up to 70 g each, one month apart, of IVIG or placebo and were followed for one year with adverse event monitoring. The primary endpoint was safety and tolerability of IVIG. Neutralizing antibody (nAb) to common BKPyV strains and BK-specific CD4+/CD8+ T-cell and natural killer cell (NKC) responses were evaluated. Results There were no adverse events. 40.0% of recipients in the treatment arm and 44.4% in the control arm cleared BK DNAemia at three months (RR 0.90; 95% CI, 0.23-2.89). Overall, 80% of recipients with a sequenced BK genotype possessed cognate nAb at baseline, and 100% acquired them by three months. Viral clearance was associated with higher percentages of BKPyV-specific CD8+ T-cells prior to IVIG (0.19% vs 3.01%, p < 0.01). Conclusions In this pilot study, two doses of IVIG were safe, but its impact on viral clearance is unclear; control of DNAemia may depend upon intrinsic virus-specific host cellular and humoral response, but further studies are needed.","PeriodicalId":501010,"journal":{"name":"The Journal of Infectious Diseases","volume":"37 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145260725","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rimke R de Kroon,Irini A M Kreulen,Mark Davids,Isabelle A M van Thiel,Iris Admiraal,Xanthe Verdoes,Mirjam M van Weissenbruch,Hendrik Niemarkt,Wouter J de Jonge,Tim de Meij,
{"title":"The gut as a source of infection for fungal pathogens: increased fecal Candida albicans precedes onset of Candida late-onset sepsis in very preterm infants.","authors":"Rimke R de Kroon,Irini A M Kreulen,Mark Davids,Isabelle A M van Thiel,Iris Admiraal,Xanthe Verdoes,Mirjam M van Weissenbruch,Hendrik Niemarkt,Wouter J de Jonge,Tim de Meij, ","doi":"10.1093/infdis/jiaf524","DOIUrl":"https://doi.org/10.1093/infdis/jiaf524","url":null,"abstract":"BACKGROUNDThe skin-to-blood route is traditionally considered the main pathway in Candida late-onset sepsis (LOS) development in preterm infants. However, emerging evidence suggests that the gut also serves as a source of infection. We aimed to characterize fecal mycobiota and microbiota profiles preceding onset of Candida LOS to assess the role of the preterm gut microbiome in disease development.METHODSIn this multicenter, case-control study, very preterm infants (<30 weeks of gestation) with Candida LOS were included. Each case was matched to non-affected controls by gestational and postnatal age, hospital site, and/or cumulative antibiotic exposure prior to day of LOS onset (t=0). Fecal samples collected at t=0 and the five preceding days were analyzed using ITS1 and 16S RNA sequencing. Microbial amplicon yields, composition, and inter-kingdom correlations were assessed.RESULTSOf 2,397 screened infants, fecal samples were available for 8/19 infants with Candida LOS. In these 8 cases, the ITS/16S amplicon yield ratio was increased (p<0.001) and the relative abundance of fecal Candida albicans correlated positively with fungal amplicon yield (ρ=0.71, padj=0.005), suggesting increased absolute abundance up to five days before onset. Additionally, bacterial yields were significantly lower (p=0.02) and α-diversity was significantly decreased (p=0.012), compared to the controls.CONCLUSIONSIncreased fecal C. albicans preceded Candida LOS onset, implicating the preterm gut as a potential source of infection. Reduced bacterial yields and diversity suggest ecological alterations that may facilitate Candida pathogenicity in the preterm gut. These findings support further research into gut-derived Candida LOS and potential for microbiota-targeted prevention strategies.","PeriodicalId":501010,"journal":{"name":"The Journal of Infectious Diseases","volume":"18 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145261445","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Fredericka Albertina Sesay,Kevin Oware,Lauren R Violette,Deborah Donnell,Josephine B Odoyo,Victor Omollo,Felix O Mogaka,R Scott McClelland,Jennifer E Balkus,Elizabeth A Bukusi,Jared M Baeten,Jenell Stewart
{"title":"Trichomonas vaginalis Among Cisgender Women Taking Doxycycline Postexposure Prophylaxis.","authors":"Fredericka Albertina Sesay,Kevin Oware,Lauren R Violette,Deborah Donnell,Josephine B Odoyo,Victor Omollo,Felix O Mogaka,R Scott McClelland,Jennifer E Balkus,Elizabeth A Bukusi,Jared M Baeten,Jenell Stewart","doi":"10.1093/infdis/jiaf519","DOIUrl":"https://doi.org/10.1093/infdis/jiaf519","url":null,"abstract":"Doxycycline treats and prevents several bacterial and parasitic infections and has been proposed as a possible treatment for Trichomonas vaginalis. Nested in a trial of doxycycline postexposure prophylaxis among cisgender women in Kenya, we conducted a secondary analysis of incident T. vaginalis infections. Among 224 cisgender women randomized to doxycycline postexposure prophylaxis and 225 to standard-of-care, there were 27 T. vaginalis diagnoses in the doxycycline postexposure prophylaxis group and 29 in the standard-of-care group (RR: 0.96, 95% CI: 0.54-1.73, p=0.9)). Understanding doxycycline postexposure prophylaxis's impact on T. vaginalis remains an important public health question that warrants further investigation.","PeriodicalId":501010,"journal":{"name":"The Journal of Infectious Diseases","volume":"36 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145246816","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Richard C White,Rachael G Lewis,James D Little,Brenda L Fredericksen,Carol L Sabourin,M Chelsea Lane,Shannon G Loelius,Kimberly A Hofmeyer,Matthew Steele,Xiaomi Tong,Robert A Johnson
{"title":"A Framework for Assessing the Opportunity for Advanced Research, Development, and Regulatory Approval of Medical Countermeasures: A Component of BARDA's Emerging Infectious Diseases Strategy.","authors":"Richard C White,Rachael G Lewis,James D Little,Brenda L Fredericksen,Carol L Sabourin,M Chelsea Lane,Shannon G Loelius,Kimberly A Hofmeyer,Matthew Steele,Xiaomi Tong,Robert A Johnson","doi":"10.1093/infdis/jiaf486","DOIUrl":"https://doi.org/10.1093/infdis/jiaf486","url":null,"abstract":"BACKGROUNDThe advanced research, development, and regulatory approval of medical countermeasures (MCMs) for emerging pathogens remains critical to national health security. We created a conceptual framework to assess the feasibility of generating pivotal data to support FDA regulatory approval of vaccines and therapeutics against known pathogens. Our framework is intended to guide key portfolio decisions on developing MCMs for emerging viral pathogens.METHODSThis framework draws on prior experience with MCM development programs, current FDA guidance, and insights from scientific, regulatory, and public health subject matter experts. We identified key requirements that impact the ability to generate pivotal data and assessed the likelihood of meeting these requirements based on current epidemiology, technical capabilities, and infrastructure. To demonstrate utility, we applied the framework to a subset of prioritized pathogens.RESULTSWe identified eight factors central to assessing the feasibility of advanced development and FDA approval of vaccines and therapeutics. These factors were used to evaluate seven emerging pathogens to illustrate how the framework may inform investment decisions across disease contexts.CONCLUSIONSThis framework supports more efficient resource allocation by highlighting MCM candidates with the highest potential for FDA approval within existing regulatory paradigms. Given current conditions, vaccine development appears more feasible than therapeutics for the pathogens assessed, although regulatory pathways remain product- and context-specific. Close consultation with the FDA will be critical in defining appropriate regulatory strategies. This framework offers a structured, proactive approach to advance MCM development and strengthen national preparedness against emerging pathogens.","PeriodicalId":501010,"journal":{"name":"The Journal of Infectious Diseases","volume":"51 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145246815","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Prevalence of candidate vaccine targets and genomic features of pediatric invasive Streptococcus agalactiae in Japan.","authors":"Masashi Kasai,Satoshi Nakano,Shota Koide,Shogo Otake,Meiwa Shibata,Kasumi Ishida-Kuroki,Yo Sugawara,Yukihiro Akeda,Kandai Nozu,Motoyuki Sugai","doi":"10.1093/infdis/jiaf491","DOIUrl":"https://doi.org/10.1093/infdis/jiaf491","url":null,"abstract":"BACKGROUNDStreptococcus agalactiae (Group B Streptococcus, GBS) is a leading cause of invasive neonatal and infant infections, including sepsis and meningitis. Despite intrapartum antibiotic prophylaxis, GBS remains a public health concern. This study aimed to estimate the vaccine coverage and characterize the genomic epidemiology of pediatric invasive GBS in Japan.METHODSWe conducted a nationwide, multicenter, retrospective genomic surveillance study involving 237 GBS isolates from sterile specimens of children aged ≤15 years across 35 hospitals in Japan between 2004 and 2023. Serotyping, antimicrobial susceptibility testing, whole-genome sequencing, and phylogenetic and single nucleotide polymorphism (SNP)-based analyses were performed.RESULTSSerotype III was the most common (48.1%), followed by Ia (27.4%) and Ib (12.2%). The estimated vaccine coverage was 98.3% for the hexavalent polysaccharide vaccine and 94.9% for the GBS-NN/NN2 protein vaccine. All isolates were susceptible to penicillin. Erythromycin and clindamycin resistance were observed in 61.2% and 43.5%, respectively. Among the 75 CC17 isolates, 59 (78.7%) contained only PI-2B and harbored both ermB and tetO, indicating the predominance of a multidrug-resistant clone. SNP-based analysis revealed evidence of nosocomial transmission and persistent regional circulation, particularly within the ST17 and ST23 lineages.CONCLUSIONThis nationwide genomic surveillance study suggests that current maternal GBS vaccine candidates would provide broad coverage for pediatric invasive infections in Japan. The identification of persistent and regionally disseminated lineages highlights the importance of investigating potential, yet poorly understood, transmission routes, including environmental reservoirs, to inform future prevention strategies.","PeriodicalId":501010,"journal":{"name":"The Journal of Infectious Diseases","volume":"19 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145246960","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Samantha Malatesta,Tara Carney,Nandi Niemand Wolhuter,Victoria Overbeck,Danie Theron,Sarah E Weber,Christina S Meade,Sarah Thomson,Tara C Bouton,Maha Farhat,Bronwyn Myers,Robin Wood,Frances Ratangee,C Robert Horsburgh,Laura F White,Robin M Warren,Karen R Jacobson
{"title":"Tuberculosis Disease Prevalence Among People Who Smoke Illicit Drugs: A Respondent- Driven Sampling Study in the Western Cape, South Africa.","authors":"Samantha Malatesta,Tara Carney,Nandi Niemand Wolhuter,Victoria Overbeck,Danie Theron,Sarah E Weber,Christina S Meade,Sarah Thomson,Tara C Bouton,Maha Farhat,Bronwyn Myers,Robin Wood,Frances Ratangee,C Robert Horsburgh,Laura F White,Robin M Warren,Karen R Jacobson","doi":"10.1093/infdis/jiaf481","DOIUrl":"https://doi.org/10.1093/infdis/jiaf481","url":null,"abstract":"BACKGROUNDTuberculosis (TB) transmission is heterogenous, yet high-risk populations remain poorly defined. We aimed to assess whether people who smoke drugs (PWSD) have elevated TB disease rates in a high-burden setting.METHODSWe recruited PWSD from a rural community in the Western Cape, South Africa, using respondent-driven sampling (RDS). Participants were ≥15 years old, tested positive for methamphetamine and/or methaqualone, and completed TB and HIV testing and biobehavioral surveys. We defined TB disease as culturable TB, Xpert MTB/RIF Ultra (Ultra) MTB detected with no history of TB, Ultra traceamong persons with HIV (PWH) >2 years from any prior diagnosis, or currently on TB treatment. We summarized population-level characteristics and estimated TB prevalence using the RDS-II estimator. We identified characteristics associated with TB using logistic regression.RESULTSBetween April 2021 and October 2023, we enrolled 750 PWSD. Overall, 71.5% (95% CI, 66.1%-76.8%) were male and the mean age was 34 years (95% CI, 33%-36%); 17.5% (95% CI, 13.0%-22.0%) were PWH, of whom 31.6% were newly diagnosed. RDS-adjusted TB prevalence was 10.4% (95% CI, 6.5%-14.3%). TB prevalence among PWSD without HIV was 8.1% (95% CI, 4.4%-11.9%), compared to 20.9% (95% CI, 8.4%-33.4%) with HIV. PWH had 3.3-fold greater adjusted odds of having TB disease (95% CI, 1.9%-5.8%).CONCLUSIONSPWSD identified through RDS had substantially elevated TB and HIV rates, with 20% of PWSD with HIV having TB. We successfully engaged PWSD in TB screening using peer recruitment. These findings highlight opportunities for community transmission identification and interventions.","PeriodicalId":501010,"journal":{"name":"The Journal of Infectious Diseases","volume":"1 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145246810","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Evaluating clinical impacts of accessory genome elements: ETT2 in Escherichia coli and beyond!","authors":"Abi Manesh,Balaji Veeraraghavan,David L Paterson","doi":"10.1093/infdis/jiaf515","DOIUrl":"https://doi.org/10.1093/infdis/jiaf515","url":null,"abstract":"","PeriodicalId":501010,"journal":{"name":"The Journal of Infectious Diseases","volume":"28 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145241108","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}