Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Maureen Durning is active.

Publication


Featured researches published by Maureen Durning.


The International Journal of Developmental Biology | 2010

On the role of placental major histocompatibility complex and decidual leukocytes in implantation and pregnancy success using non-human primate models

Thaddeus G. Golos; Gennadiy I. Bondarenko; Svetlana V. Dambaeva; Edith E. Breburda; Maureen Durning

While there is broad agreement that interactions of the human maternal immune system with the tissues and cells of the implanting embryo are likely to be critical contributors to pregnancy success, there remains a dearth of information which directly confirms this expectation. Although animal models of reproductive function often provide opportunities for confirming such hypotheses, progress in this area has been sporadic due to limitations of traditional laboratory or agricultural animal models, such as rodents, sheep, pigs and cattle. Many of these limitations derive from divergent modes of implantation and placentation across mammalian species. Over the past decade there has been progress in the development of the nonhuman primate as a model in which to address questions of pregnancy success in the area of immunology. The purpose of this review is to compare available model species, summarize current knowledge and recent progress with an emphasis on experimental in vivo manipulations, and suggest areas available for additional study and growth.


Journal of Immunology | 2007

Passive Immunization against the MHC Class I Molecule Mamu-AG Disrupts Rhesus Placental Development and Endometrial Responses

Gennadiy I. Bondarenko; David W. Burleigh; Maureen Durning; Edith E. Breburda; Richard L. Grendell; Thaddeus G. Golos

The unique MHC phenotype of the human and nonhuman primate placenta has suggested a potential role in maternal-fetal immune tolerance, pregnancy success, and maternal as well as fetal well-being. In the rhesus monkey (Macaca mulatta) a nonclassical MHC class I molecule, Mamu-AG, is a putative homologue of HLA-G and is hypothesized to play a role in maternal-fetal immune interactions during pregnancy. Rhesus monkeys were passively immunized during the second week after implantation with a mAb against Mamu-AG. Passive immunization altered the growth and vascularization of the fetal placenta, the placental modification of maternal endometrial vessels, the maternal leukocyte response to implantation, and the differentiation of epithelial and stromal cells in the endometrium. These data are the first to demonstrate in vivo the importance of MHC class I molecules expressed on primate trophoblasts in establishing an important environment for pregnancy success through coordinated interactions between endometrial and fetal tissues.


Biology of Reproduction | 2012

Immunophenotype and Cytokine Profiles of Rhesus Monkey CD56bright and CD56dim Decidual Natural Killer Cells

Svetlana V. Dambaeva; Maureen Durning; Ann E. Rozner; Thaddeus G. Golos

ABSTRACT The primate endometrium is characterized in pregnancy by a tissue-specific population of CD56bright natural killer (NK) cells. These cells are observed in human, rhesus, and other nonhuman primate decidua. However, other subsets of NK cells are present in the decidua and may play distinct roles in pregnancy. The purpose of this study was to define the surface marker phenotype of rhesus monkey decidual NK (dNK) cell subsets, and to address functional differences by profiling cytokine and chemokine secretion in contrast with decidual T cells and macrophages. Rhesus monkey decidual leukocytes were obtained from early pregnancy tissues, and were characterized by flow cytometry and multiplex assay of secreted factors. We concluded that the major NK cell population in rhesus early pregnancy decidua are CD56bright CD16+NKp30− decidual NK cells, with minor CD56dim and CD56neg dNK cells. Intracellular cytokine staining demonstrated that CD56dim and not CD56bright dNK cells are the primary interferon-gamma (IFNG) producers. In addition, the profile of other cytokines, chemokines, and growth factors secreted by these two dNK cell populations was generally similar, but distinct from that of peripheral blood NK cells. Finally, analysis of multiple pregnancies from eight dams revealed that the decidual immune cell profile is characteristic of an individual animal and is consistently maintained across successive pregnancies, suggesting that the uterine immune environment in pregnancy is carefully regulated in the rhesus monkey decidua.


Molecular and Cellular Endocrinology | 2004

Id2 Is a primary partner for the E2-2 basic helix-loop-helix transcription factor in the human placenta

Yi-Ping Liu; David W. Burleigh; Maureen Durning; Lori Hudson; Ing-Ming Chiu; Thaddeus G. Golos

We screened a term placental cDNA library by the yeast two-hybrid approach with Id2, a negative regulator of basic helix-loop-helix (bHLH) factors. Of the clones obtained, approximately one-third were the E2-2 bHLH transcription factor. Id2 and E2-2 were shown to interact in direct two-hybrid assays in yeast cells, as well as immunoprecipitation assays in mammalian cells. Immunohistochemical analysis demonstrated co-localization of both Id2 and E2-2 in placental trophoblasts. Co-transfection of JEG-3 cells with E2-2 and Id2, and a luciferase reporter construct under the control of the human chorionic gonadotropin alpha-subunit promoter revealed that E2-2 had a negative effect on CGalpha-subunit transcription, which could be relieved by overexpression of Id2. The library was in turn rescreened with E2-2, and Id2 and Id1 were essentially the only clones obtained. We conclude that Id2 is a primary binding partner for the bHLH transcription factor E2-2 in the human placenta.


American Journal of Reproductive Immunology | 2011

Modulation of Cytokine and Chemokine Secretions in Rhesus Monkey Trophoblast Co-Culture With Decidual but not Peripheral Blood Monocyte–Derived Macrophages

Ann E. Rozner; Svetlana V. Dambaeva; Jessica G. Drenzek; Maureen Durning; Thaddeus G. Golos

Citation 
Rozner AE, Dambaeva SV, Drenzek JG, Durning M, Golos TG. Modulation of cytokine and chemokine secretions in rhesus monkey trophoblast co‐culture with decidual but not peripheral blood monocyte–derived macrophages. Am J Reprod Immunol 2011; 66: 115–127


Journal of Reproductive Immunology | 2009

Characterization of decidual leukocyte populations in cynomolgus and vervet monkeys.

Svetlana V. Dambaeva; Edith E. Breburda; Maureen Durning; Mark A. Garthwaite; Thaddeus G. Golos

The objective of this study was the phenotypic and functional evaluation of decidual immune cells in the cynomolgus and vervet monkeys. Early pregnancy (days 36-42) deciduas were obtained by fetectomy for histological evaluation and decidual mononuclear leukocyte (MNL) isolation. While peripheral NK (pNK) cells in these species do not express CD56, CD56(+) NK cells were abundant in decidual samples. The majority of decidual NK (dNK) cells (>80%) had high light-scatter characteristics and were CD56(bright)CD16(+) cells with no or very low levels of natural cytotoxicity receptors (NKp46, NKp30) and NKG2A, while a minor population were small CD56(dim)CD16(-) lymphocytes also expressing less NKp46, NKp30 and NKG2A than pNK cells. All dNK cells were found to be perforin(+); however, their cytotoxic potential was low and cynomolgus dNK cells showed strongly reduced cytotoxicity against target cells compared with pNK cells. Macrophages and T cells together comprised approximately 25-30% of decidual MNL. Decidual T cells contained a higher proportion of the minor T cell subtypes (gammadeltaT cells, CD56(+) T cells) compared with peripheral blood. A subset of DC-SIGN(+) macrophages, with a distribution adjacent to areas of placental attachment in contrast to the widespread setting of general CD68(+) cells, was identified in both species. Together, these results demonstrate that the maternal-fetal interface in both cynomolgus and vervet monkeys is very rich in immune cells that have similar phenotypes to those seen in humans, indicating that both species are excellent models to study the contributions of distinct immune cell populations to pregnancy support.


Mbio | 2017

Acute Fetal Demise with First Trimester Maternal Infection Resulting from Listeria monocytogenes in a Nonhuman Primate Model

Bryce Wolfe; Gregory J. Wiepz; Michele L. Schotzko; Gennadiy I. Bondarenko; Maureen Durning; Heather A. Simmons; Andres Mejia; Nancy G. Faith; Emmanuel Sampene; M. Suresh; Sophia Kathariou; Charles J. Czuprynski; Thaddeus G. Golos

ABSTRACT Infection with Listeria monocytogenes during pregnancy is associated with miscarriage, preterm birth, and neonatal complications, including sepsis and meningitis. While the risk of these conditions is thought to be greatest during the third trimester of pregnancy, the determinants of fetoplacental susceptibility to infection, the contribution of gestational age, and the in vivo progression of disease at the maternal-fetal interface are poorly understood. We developed a nonhuman primate model of listeriosis to better understand antecedents of adverse pregnancy outcomes in early pregnancy. Four pregnant cynomolgus macaques (Macaca fascicularis) received a single intragastric inoculation between days 36 and 46 of gestation with 107 CFU of an L. monocytogenes strain isolated from a previous cluster of human listeriosis cases that resulted in adverse pregnancy outcomes. Fecal shedding, maternal bacteremia, and fetal demise were consistently noted within 7 to 13 days. Biopsy specimens of maternal liver, spleen, and lymph node displayed variable inflammation and relatively low bacterial burden. In comparison, we observed greater bacterial burden in the decidua and placenta and the highest burden in fetal tissues. Histopathology indicated vasculitis, fibrinoid necrosis, and thrombosis of the decidual spiral arteries, acute chorioamnionitis and villitis in the placenta, and hematogenous infection of the fetus. Vascular pathology suggests early impact of L. monocytogenes infection on spiral arteries in the decidua, which we hypothesize precipitates subsequent placentitis and fetal demise. These results demonstrate that L. monocytogenes tropism for the maternal reproductive tract results in infection of the decidua, placenta, and the fetus itself during the first trimester of pregnancy. IMPORTANCE Although listeriosis is known to cause significant fetal morbidity and mortality, it is typically recognized in the third trimester of human pregnancy. Its impact on early pregnancy is poorly defined. Here we provide evidence that exposure to L. monocytogenes in the first trimester poses a greater risk of fetal loss than currently appreciated. Similarities in human and nonhuman primate placentation, physiology, and reproductive immunology make this work highly relevant to human pregnancy. We highlight the concept that the maternal immune response that protects the mother from serious disease is unable to protect the fetus, a concept relevant to classic TORCH (toxoplasmosis, other, rubella, cytomegalovirus, and herpes) infections and newly illuminated by current Zika virus outbreaks. Studies with this model, using the well-understood organism L. monocytogenes, will permit precise analysis of host-pathogen interactions at the maternal-fetal interface and have broad significance to both recognized and emerging infections in the setting of pregnancy. Although listeriosis is known to cause significant fetal morbidity and mortality, it is typically recognized in the third trimester of human pregnancy. Its impact on early pregnancy is poorly defined. Here we provide evidence that exposure to L. monocytogenes in the first trimester poses a greater risk of fetal loss than currently appreciated. Similarities in human and nonhuman primate placentation, physiology, and reproductive immunology make this work highly relevant to human pregnancy. We highlight the concept that the maternal immune response that protects the mother from serious disease is unable to protect the fetus, a concept relevant to classic TORCH (toxoplasmosis, other, rubella, cytomegalovirus, and herpes) infections and newly illuminated by current Zika virus outbreaks. Studies with this model, using the well-understood organism L. monocytogenes, will permit precise analysis of host-pathogen interactions at the maternal-fetal interface and have broad significance to both recognized and emerging infections in the setting of pregnancy.


American Journal of Reproductive Immunology | 2012

Immunomorphological Changes in the Rhesus Monkey Endometrium and Decidua During the Menstrual Cycle and Early Pregnancy

Gennadiy I. Bondarenko; Maureen Durning; Thaddeus G. Golos

Throughout the reproductive cycle and into early pregnancy, the normal endometrium undergoes changes in a range of leukocytes, epithelia, stromal fibroblasts, and vascular structures caused by intersecting effects of hormone balance and embryo implantation. The direct investigation in humans of reproductive tract responses during normal and physiologically altered cycles is not practical or feasible.


Journal of Immunological Methods | 2009

Generation of macrophages from peripheral blood monocytes in the rhesus monkey

Ann E. Rozner; Svetlana V. Dambaeva; Jessica G. Drenzek; Maureen Durning; Thaddeus G. Golos

Macrophages are found in tissues throughout the body and are important immune cells, however, these tissue macrophages are difficult to collect and study. Therefore, the ability to differentiate macrophages from peripheral blood precursors is an important research tool. Macrophage differentiation has been well studied in humans, but differentiation in the non-human primate is poorly characterized. Using human models is not always feasible for invasive experimental studies and, therefore, developing reliable protocols for the non-human primate model is important. We describe a method to differentiate macrophages in vitro in the rhesus monkey by culturing adherent peripheral blood mononuclear cells for five days in RPMI-1640 supplemented with 1% human serum, M-CSF, and IL-1beta. The resulting cells had a distinct macrophage phenotype, the ability to secrete cytokines in response to LPS, and antigen uptake and processing capabilities.


Immunogenetics | 2009

Characterization of cynomolgus and vervet monkey placental MHC class I expression: diversity of the nonhuman primate AG locus

Gennadiy I. Bondarenko; Svetlana V. Dambaeva; Richard L. Grendell; Austin L. Hughes; Maureen Durning; Mark A. Garthwaite; Thaddeus G. Golos

Nonhuman primates are important animal models for the study of the maternal immune response to implantation within the decidua. The objective of this study was to define the placental expression of major histocompatibility complex (MHC) class I molecules in the cynomolgus (Macaca fascicularis) and vervet (African green) (Chlorocebus aethiops) monkeys. Early pregnancy (d36-42) cynomolgus and vervet placentas were obtained by fetectomy and prepared for histological evaluation. A pan-MHC class I monoclonal antibody demonstrated MHC class I expression in both vervet and cynomolgus placental trophoblasts, with particularly high expression in the villous syncytium, as previously shown in the rhesus and baboon. Placental cytotrophoblasts were isolated by enzymatic dispersion and gradient centrifugation and cultured, and multicolor flow cytometry was used to phenotype cell populations. Culture of isolated villous cytotrophoblasts demonstrated that MHC class I expression was linked to syncytiotrophoblast differentiation. A monoclonal antibody against Mamu-AG, the nonclassical MHC class I homolog of HLA-G in the rhesus monkey, demonstrated intense immunostaining and cell surface expression in cynomolgus placental trophoblasts; however, staining with vervet placenta and cells was low and inconsistent. Reverse transcriptase polymerase chain reaction was used to clone MHC class I molecules expressed in cynomolgus and vervet placentas. While Mafa-AG messenger RNA (mRNA) was readily detectable in cynomolgus placental RNA and was >99% identical at the amino acid level with Mamu-AG, 7/8 Chae-AG complementary DNAs had an unusual 16 amino acid repeat in the α1 domain, and all clones had an unexpected absence of the early stop codon at the 3′-end of the mRNA diagnostic for rhesus, cynomolgus, and baboon AG mRNAs, as well as HLA-G. We conclude that while the vervet monkey has retained the placental expression of a primate-specific nonclassical MHC class I locus, diversity is also revealed in this locus expressed at the maternal–fetal interface, thought to participate in placental regulation of the maternal immune response to embryo implantation and pregnancy.

Collaboration


Dive into the Maureen Durning's collaboration.

Top Co-Authors

Avatar

Thaddeus G. Golos

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Svetlana V. Dambaeva

Rosalind Franklin University of Medicine and Science

View shared research outputs
Top Co-Authors

Avatar

Gennadiy I. Bondarenko

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jessica G. Drenzek

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Richard L. Grendell

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Ann E. Rozner

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Donald J. Dierschke

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Edith E. Breburda

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jennifer M. Fisher

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Mark A. Garthwaite

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge