Your browser doesn't support javascript.
loading
Relationship of maternal cytomegalovirus-specific antibody responses and viral load to vertical transmission risk following primary maternal infection in a rhesus macaque model.
Otero, Claire E; Barfield, Richard; Scheef, Elizabeth; Nelson, Cody S; Rodgers, Nicole; Wang, Hsuan-Yuan; Moström, Matilda J; Manuel, Tabitha D; Sass, Julian; Schmidt, Kimberli; Taher, Husam; Papen, Courtney; Sprehe, Lesli; Kendall, Savannah; Davalos, Angel; Barry, Peter A; Früh, Klaus; Pollara, Justin; Malouli, Daniel; Chan, Cliburn; Kaur, Amitinder; Permar, Sallie R.
  • Otero CE; Department of Pathology, Duke University, Durham, NC.
  • Barfield R; Department of Pediatrics, Weill Cornell Medical College, New York, NY.
  • Scheef E; Department of Biostatistics and Bioinformatics, Duke University, Durham, NC.
  • Nelson CS; Tulane National Primate Research Center, Covington, LA.
  • Rodgers N; Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Boston, MA.
  • Wang HY; Duke Human Vaccine Institute & Department of Surgery, Duke University, Durham, NC.
  • Moström MJ; Department of Pediatrics, Weill Cornell Medical College, New York, NY.
  • Manuel TD; Department of Immunology, Duke University, Durham, NC.
  • Sass J; Tulane National Primate Research Center, Covington, LA.
  • Schmidt K; Tulane National Primate Research Center, Covington, LA.
  • Taher H; Department of Mathematics, North Carolina State University, Raleigh, NC.
  • Papen C; Center for Immunology and Infectious Diseases, University of California, Davis, CA.
  • Sprehe L; Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR.
  • Kendall S; Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR.
  • Davalos A; Tulane National Primate Research Center, Covington, LA.
  • Barry PA; Tulane National Primate Research Center, Covington, LA.
  • Früh K; Department of Biostatistics and Bioinformatics, Duke University, Durham, NC.
  • Pollara J; Center for Immunology and Infectious Diseases, University of California, Davis, CA.
  • Malouli D; Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR.
  • Chan C; Duke Human Vaccine Institute & Department of Surgery, Duke University, Durham, NC.
  • Kaur A; Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR.
  • Permar SR; Department of Biostatistics and Bioinformatics, Duke University, Durham, NC.
bioRxiv ; 2023 Apr 21.
Article en En | MEDLINE | ID: mdl-37131785
ABSTRACT
Cytomegalovirus (CMV) is the most common congenital infection and cause of birth defects worldwide. Primary CMV infection during pregnancy leads to a higher frequency of congenital CMV (cCMV) than maternal re-infection, suggesting that maternal immunity confers partial protection. However, poorly understood immune correlates of protection against placental transmission contributes to the current lack of an approved vaccine to prevent cCMV. In this study, we characterized the kinetics of maternal plasma rhesus CMV (RhCMV) viral load (VL) and RhCMV-specific antibody binding and functional responses in a group of 12 immunocompetent dams with acute, primary RhCMV infection. We defined cCMV transmission as RhCMV detection in amniotic fluid (AF) by qPCR. We then leveraged a large group of past and current primary RhCMV infection studies in late-first/early-second trimester RhCMV-seronegative rhesus macaque dams, including immunocompetent (n=15), CD4+ T cell-depleted with (n=6) and without (n=6) RhCMV-specific polyclonal IgG infusion before infection to evaluate differences between RhCMV AF-positive and AF-negative dams. During the first 3 weeks after infection, the magnitude of RhCMV VL in maternal plasma was higher in AF-positive dams in the combined cohort, while RhCMV glycoprotein B (gB)- and pentamer-specific binding IgG responses were lower magnitude compared to AF-negative dams. However, these observed differences were driven by the CD4+ T cell-depleted dams, as there were no differences in plasma VL or antibody responses between immunocompetent AF-positive vs AF-negative dams. Overall, these results suggest that levels of neither maternal plasma viremia nor humoral responses are associated with cCMV following primary maternal infection in healthy individuals. We speculate that other factors related to innate immunity are more important in this context as antibody responses to acute infection likely develop too late to influence vertical transmission. Yet, pre-existing CMV glycoprotein-specific and neutralizing IgG may provide protection against cCMV following primary maternal CMV infection even in high-risk, immunocompromised settings.

Texto completo: 1 Banco de datos: MEDLINE Tipo de estudio: Etiology_studies / Prognostic_studies / Risk_factors_studies Idioma: En Año: 2023 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Tipo de estudio: Etiology_studies / Prognostic_studies / Risk_factors_studies Idioma: En Año: 2023 Tipo del documento: Article