Your browser doesn't support javascript.
loading
Innate and Adaptive Immune Parameters following mRNA Vaccination in Mice.
Bonam, Srinivasa Reddy; Hazell, Nicholas C; Mathew, Mano Joseph; Liang, Yuejin; Zhang, Xuxiang; Wei, Zhi; Alameh, Mohamad-Gabriel; Weissman, Drew; Hu, Haitao.
Afiliación
  • Bonam SR; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
  • Hazell NC; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
  • Mathew MJ; Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
  • Liang Y; EFREI Research Lab, Panthéon Assas University, 30-32 Avenue de la République, 94800 Villejuif, France.
  • Zhang X; Laboratoire Génomique, Bioinformatique et Chimie Moléculaire, EA7528, Conservatoire National des Arts et Métiers, HESAM Université, 2 rue Conté, 75003 Paris, France.
  • Wei Z; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
  • Alameh MG; Department of Computer Science, New Jersey Institute of Technology, Newark, NJ 07102, USA.
  • Weissman D; Department of Computer Science, New Jersey Institute of Technology, Newark, NJ 07102, USA.
  • Hu H; Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA.
Vaccines (Basel) ; 12(5)2024 May 15.
Article en En | MEDLINE | ID: mdl-38793794
ABSTRACT
The COVID-19 pandemic has raised the standard regarding the current vaccine development pace, as several messenger RNA (mRNA)-lipid nanoparticle (LNP) vaccines have proved their ability to induce strong immunogenicity and protective efficacy. We developed 1-methylpseudouridine-containing mRNA-LNP vaccines, expressing either the more conserved SARS-CoV-2 nucleoprotein (mRNA-N) or spike protein (mRNA-S), both based on the prototypic viral sequences. When combining both mRNA-S and mRNA-N together (mRNA-S+N), the vaccine showed high immunogenicity and broad protection against different SARS-CoV-2 variants, including wildtype, Delta, BA.1, BA.5, and BQ.1. To better understand the mechanisms behind this broad protection obtained by mRNA-S+N, we analyzed innate and adaptive immune parameters following vaccination in mice. Compared to either mRNA-S or mRNA-N alone, mice vaccinated with mRNA-S+N exhibited an increase in the innate immune response, as depicted by the higher cytokine (IL-6 and chemokine (MCP-1) levels. In addition, lymph node immunophenotyping showed the maturation and activation of dendritic cells and natural killer cells, respectively. To understand the adaptive immune response, RNA-Seq analyses of the lung and spleen samples of the vaccinated mice were performed in parallel and revealed a stronger immune gene-expression profile in the lung than that in the spleen. Compared to mRNA-S alone, mRNA-S+N vaccination elicited higher levels of expression for genes involved in multiple immune pathways, including T cells, cytokine signaling, antigen presentation, B cells, and innate immunity. Together, our studies provide immunological insights into the mechanisms of broad protection conferred by dual mRNA vaccination against SARS-CoV-2 variants.
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Idioma: En Revista: Vaccines (Basel) Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Idioma: En Revista: Vaccines (Basel) Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos