Your browser doesn't support javascript.
loading
Primate innate immune responses to bacterial and viral pathogens reveals an evolutionary trade-off between strength and specificity.
Hawash, Mohamed B F; Sanz-Remón, Joaquin; Grenier, Jean-Christophe; Kohn, Jordan; Yotova, Vania; Johnson, Zach; Lanford, Robert E; Brinkworth, Jessica F; Barreiro, Luis B.
Afiliação
  • Hawash MBF; Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, QC H3T 1C5, Montréal, Canada.
  • Sanz-Remón J; Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt.
  • Grenier JC; Departamento de Fiscia Teórica, Universidad de Zaragoza, 50001, Zaragoza, Spain.
  • Kohn J; Institute for Biocomputation and Physics of Complex Systems, Universidad de Zaragoza, 50018, Zaragoza, Spain.
  • Yotova V; Montreal Heart Institute, University of Montreal, QC H1T 1C8, Montreal, Canada.
  • Johnson Z; Department of Neuroscience, Emory University, Atlanta, GA 30329.
  • Lanford RE; Department of Psychiatry, College of Health Sciences, University of California, San Diego, CA 92093.
  • Brinkworth JF; Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, QC H3T 1C5, Montréal, Canada.
  • Barreiro LB; Illumina, San Diego, CA 92122.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article em En | MEDLINE | ID: mdl-33771921
ABSTRACT
Despite their close genetic relatedness, apes and African and Asian monkeys (AAMs) differ in their susceptibility to severe bacterial and viral infections that are important causes of human disease. Such differences between humans and other primates are thought to be a result, at least in part, of interspecies differences in immune response to infection. However, because of the lack of comparative functional data across species, it remains unclear in what ways the immune systems of humans and other primates differ. Here, we report the whole-genome transcriptomic responses of ape species (human and chimpanzee) and AAMs (rhesus macaque and baboon) to bacterial and viral stimulation. We find stark differences in the responsiveness of these groups, with apes mounting a markedly stronger early transcriptional response to both viral and bacterial stimulation, altering the transcription of ∼40% more genes than AAMs. Additionally, we find that genes involved in the regulation of inflammatory and interferon responses show the most divergent early transcriptional responses across primates and that this divergence is attenuated over time. Finally, we find that relative to AAMs, apes engage a much less specific immune response to different classes of pathogens during the early hours of infection, up-regulating genes typical of anti-viral and anti-bacterial responses regardless of the nature of the stimulus. Overall, these findings suggest apes exhibit increased sensitivity to bacterial and viral immune stimulation, activating a broader array of defense molecules that may be beneficial for early pathogen killing at the potential cost of increased energy expenditure and tissue damage.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Bactérias / Vírus / Metabolismo Energético / Interações Hospedeiro-Patógeno / Imunidade Inata Limite: Adult / Animals / Female / Humans / Male / Middle aged Idioma: En Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Bactérias / Vírus / Metabolismo Energético / Interações Hospedeiro-Patógeno / Imunidade Inata Limite: Adult / Animals / Female / Humans / Male / Middle aged Idioma: En Ano de publicação: 2021 Tipo de documento: Article