Group B streptococcus activates transcriptomic pathways related to premature birth in human extraplacental membranes in vitro,

HR Park, SM Harris, E Boldenow… - Biology of …, 2018 - academic.oup.com
HR Park, SM Harris, E Boldenow, RC McEachin, M Sartor, M Chames, R Loch-Caruso
Biology of Reproduction, 2018academic.oup.com
Streptococcus agalactiae (group B streptococcus [GBS]) infection in pregnant women is the
leading cause of infectious neonatal morbidity and mortality in the United States. Although
inflammation during infection has been associated with preterm birth, the contribution of
GBS to preterm birth is less certain. Moreover, the early mechanisms by which GBS interacts
with the gestational tissue to affect adverse pregnancy outcomes are poorly understood. We
hypothesized that short-term GBS inoculation activates pathways related to inflammation …
Abstract
Streptococcus agalactiae (group B streptococcus [GBS]) infection in pregnant women is the leading cause of infectious neonatal morbidity and mortality in the United States. Although inflammation during infection has been associated with preterm birth, the contribution of GBS to preterm birth is less certain. Moreover, the early mechanisms by which GBS interacts with the gestational tissue to affect adverse pregnancy outcomes are poorly understood. We hypothesized that short-term GBS inoculation activates pathways related to inflammation and premature birth in human extraplacental membranes. We tested this hypothesis using GBS-inoculated human extraplacental membranes in vitro. In agreement with our hypothesis, a microarray-based transcriptomics analysis of gene expression changes in GBS-inoculated membranes revealed that GBS activated pathways related to inflammation and preterm birth with significant gene expression changes occurring as early as 4 h postinoculation. In addition, pathways related to DNA replication and repair were downregulated with GBS treatment. Conclusions based on our transcriptomics data were further supported by responses of prostaglandin E2 (PGE2), and matrix metalloproteinases 1 (MMP1) and 3 (MMP3), all of which are known to be involved in parturition and premature rupture of membranes. These results support our initial hypothesis and provide new information on molecular targets of GBS infection in human extraplacental membranes.
Oxford University Press