Graduation Year

2018

Document Type

Dissertation

Degree

Ph.D.

Degree Name

Doctor of Philosophy (Ph.D.)

Degree Granting Department

Medical Sciences

Major Professor

Hana Totary-Jain, Ph.D.

Committee Member

Bruce Lindsey, Ph.D.

Committee Member

Jay Dean, Ph.D.

Committee Member

Narasaiah Kolliputi, Ph.D.

Keywords

trophoblast, hypoxia, invasion, DOHaD

Abstract

An essential event during early pregnancy is the invasion of trophoblasts into the maternal decidua, which is necessary for proper implantation and establishment of maternal-fetal interface and ultimately allows for proper nutrient exchange and immunological tolerance of the growing fetus. For this invasion to occur, cells originating from the trophectoderm undergo an epithelial to mesenchymal transition to become invasive extravillous trophoblasts and begin invading the uterine decidual tissue. Through the secretion of matrix metalloproteinases and through interactions with many cytokines and cell-adhesion molecules, this well-orchestrated process of trophoblast invasion results in extensive remodeling of the maternal spiral vasculature by the extravillous trophoblasts. Ultimately, the spiral arteries are transformed from high resistance, low flow vessels to low resistance, high flow vessels to allow for adequate perfusion of the placenta and developing fetus.

Preeclampsia is a leading cause of maternal morbidity worldwide and is associated with the onset of hypertension and proteinuria, typically after 20 weeks of gestation. While the hypertension typically resolves following delivery of the fetus and placenta, both the mother and growing child are faced with long-term adverse health effects such as the development of cardiovascular disease and metabolic disorders. Preeclampsia is characterized by widespread maternal inflammation and endothelial dysfunction triggered by the secretion of soluble factors from the placenta into the maternal circulation. It is thought that the onset of these adverse systemic conditions is initiated by poor placental perfusion and pathologically hypoxic conditions in the placenta. In many cases of preeclampsia, there is evidence of shallow trophoblast invasion which results in incomplete spiral artery transformation, ultimately leading to poor placental perfusion. However, the exact mechanisms underlying the inadequate extravillous trophoblast invasion and remodeling are incompletely understood.

LIN28 is an RNA binding protein that is highly expressed in embryonic stem cells, fetal tissues and many cancers, and was discovered as a regulator of the maturation of the Let-7 family of miRNAs. However, as an RNA binding protein, LIN28 has been shown to interact with thousands of mRNA transcripts, leading to both increased and decreased protein expression, and control of many cellular processes such as differentiation, proliferation, migration, invasion, and cellular metabolism. In vertebrates, LIN28 exists as two highly homologous paralogs, LIN28A and LIN28B, however LIN28B is slightly larger and contains a nuclear localization signal not found in LIN28A. While they both function to inhibit Let-7 maturation, there is evidence to suggest they also have independent functions.

Given the primary role of LIN28A and LIN28B in modulating cell metabolism, differentiation and invasion, we hypothesized that LIN28A and/or LIN28B regulates trophoblast differentiation and invasion, and that its dysregulation may contribute to preeclampsia. We found that LIN28B mRNA expression is ~1300-fold higher than LIN28A in human term placenta and is the predominant paralog expressed in human primary cytotrophoblasts, syncytiotrophoblasts, and decidual cells. We also found that LIN28B mRNA and protein levels are significantly reduced in human placentas from preeclamptic pregnancies compared to placentas from normal pregnancies, while LIN28A expression is unchanged. Upon investigation of human first trimester placenta sections, we found that LIN28B is more highly expressed in the invasive extravillous trophoblasts and syncytial sprouts compared to villous trophoblasts. To support this with in vitro evidence, we found that overexpression of LIN28B in human HTR8/SVneo cells resulted in increased proliferation, migration and invasion, while knockdown of LIN28B in JEG3 cells resulted in decreased proliferation. Furthermore, knockdown of LIN28B in JEG3 cells led to decreased expression of SYN-1, ELABELA, and the chromosome 19 miRNA cluster, with accompanying increases in the pro-inflammatory cytokine TNFα and ITGβ4, an integrin enriched on non-invasive trophoblasts. Moreover, culture of JEG3 and BEWO cells in hypoxia resulted in significantly decreased levels of LIN28B mRNA and protein expression, as well as syncytin-1 and ELABELA mRNA levels, while TNFα was increased. These results provide the first evidence that LIN28B is the predominant paralog expressed in human placenta and decreased LIN28B may play a crucial role in PE pathogenesis by reducing trophoblast invasion, syncytialization and by promoting inflammation.

Share

COinS