Network


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

Hotspot


Dive into the research topics where Gennadiy I. Bondarenko is active.

Publication


Featured researches published by Gennadiy I. Bondarenko.


Cell Stem Cell | 2010

Pax6 Is a Human Neuroectoderm Cell Fate Determinant

Xiaoqing Zhang; Cindy Tzu-Ling Huang; Jing Chen; Matthew T. Pankratz; Jiajie Xi; Jin Li; Ying Yang; Timothy M. LaVaute; Xue Jun Li; Melvin Ayala; Gennadiy I. Bondarenko; Zhong Wei Du; Ying Jin; Thaddeus G. Golos; Su-Chun Zhang

The transcriptional regulation of neuroectoderm (NE) specification is unknown. Here we show that Pax6 is uniformly expressed in early NE cells of human fetuses and those differentiated from human embryonic stem cells (hESCs). This is in contrast to the later expression of Pax6 in restricted mouse brain regions. Knockdown of Pax6 blocks NE specification from hESCs. Overexpression of either Pax6a or Pax6b, but not Pax6triangle upPD, triggers hESC differentiation. However, only Pax6a converts hESCs to NE. In contrast, neither loss nor gain of function of Pax6 affects mouse NE specification. Both Pax6a and Pax6b bind to pluripotent gene promoters but only Pax6a binds to NE genes during human NE specification. These findings indicate that Pax6 is a transcriptional determinant of the human NE and suggest that Pax6a and Pax6b coordinate with each other in determining the transition from pluripotency to the NE fate in human by differentially targeting pluripotent and NE genes.


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.


Journal of Reproductive Immunology | 2008

Expression of indoleamine 2,3-dioxygenase in the rhesus monkey and common marmoset

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

Indoleamine 2,3-dioxygenase (IDO) catalyzes the initial and rate-limiting step of tryptophan degradation along the kynurenine pathway, and is hypothesized to limit tryptophan availability at embryo implantation and prevent maternal T cell activation at the maternal-fetal interface. To determine if nonhuman primates are suitable models for investigating the role of IDO during pregnancy, we defined the expression of IDO in the rhesus monkey and common marmoset with particular attention to the female reproductive tract and placenta. IDO mRNA was detected by RT-PCR in the rhesus monkey term placenta, lung, small intestine, spleen, lymph node and nonpregnant uterus, and also in the common marmoset placenta. Immunohistochemical analysis of rhesus monkey tissues localized IDO to glandular epithelium of nonpregnant endometrium and first trimester decidua, vessel endothelium of nonpregnant myometrium, first trimester decidua and term decidua, and villous vessel endothelium and syncytiotrophoblast of term placenta. Western blot analysis confirmed IDO in rhesus monkey term placenta. In the common marmoset, IDO was detected in glandular epithelium of the nonpregnant uterus and in the decidua at day 60 and day 128 of gestation. IDO activity was higher in rhesus monkey and common marmoset decidua and placentas than in other tissues. Confirmation of IDO expression in rhesus monkey and common marmoset uterine and placental tissues supports the hypothesis that this enzyme regulates immune activation at the maternal-fetal interface and demonstrates that nonhuman primates may provide models with distinct similarities to human placentation to study the role of IDO in maternal-fetal immune dialogue.


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.


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.


Methods in molecular medicine | 2006

Immune and Trophoblast Cells at the Rhesus Monkey Maternal-Fetal Interface

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

To promote the use of the nonhuman primate model for the study of the cellular and molecular biology of maternal-fetal interactions and placental development during early pregnancy, we have developed protocols for the isolation and characterization of placental trophoblasts and decidual immune cells from the rhesus monkey. In this chapter, we provide protocols for trophoblast and decidual immune cell isolation, phenotyping of isolated cells by flow cytometry, and analysis of placental and decidual tissues by immunohistochemistry. Information on antibodies for these analyses are also provided, which is an important consideration when attempting to use anti-human antibodies for the study of nonhuman primates.


Reproductive Biology and Endocrinology | 2018

Trophoblast differentiation, invasion and hormone secretion in a three-dimensional in vitro implantation model with rhesus monkey embryos

T. Arthur Chang; Gennadiy I. Bondarenko; Behzad Gerami-Naini; Jessica G. Drenzek; Maureen Durning; Mark A. Garthwaite; Jenna Kropp Schmidt; Thaddeus G. Golos

BackgroundThe initiation of primate embryo invasion into the endometrium and the formation of the placenta from trophoblasts, fetal mesenchyme, and vascular components are essential for the establishment of a successful pregnancy. The mechanisms which direct morphogenesis of the chorionic villi, and the interactions between trophectoderm-derived trophoblasts and the fetal mesenchyme to direct these processes during placentation are not well understood due to a dearth of systems to examine and manipulate real-time primate implantation. Here we describe an in vitro three-dimensional (3-D) model to study implantation which utilized IVF-generated rhesus monkey embryos cultured in a Matrigel explant system.MethodsBlastocyst stage embryos were embedded in a 3-D microenvironment of a Matrigel carrier and co-cultured with a feeder layer of cells generating conditioned medium. Throughout the course of embryo co-culture embryo growth and secretions were monitored. Embedded embryos were then sectioned and stained for markers of trophoblast function and differentiation.ResultsSigns of implantation were observed including enlargement of the embryo mass, and invasion and proliferation of trophoblast outgrowths. Expression of chorionic gonadotropin defined by immunohistochemical staining, and secretion of chorionic gonadotropin and progesterone coincident with the appearance of trophoblast outgrowths, supported the conclusion that a trophoblast cell lineage formed from implanted embryos. Positive staining for selected markers including Ki67, MHC class I, NeuN, CD31, vonWillebrand Factor and Vimentin, suggest growth and differentiation of the embryo following embedding.ConclusionsThis 3-D in vitro system will facilitate further study of primate embryo biology, with potential to provide a platform for study of genes related to implantation defects and trophoblast differentiation.


Placenta | 2008

Non-classical MHC-E (Mamu-E) expression in the rhesus monkey placenta.

Svetlana V. Dambaeva; Gennadiy I. Bondarenko; Richard L. Grendell; Rachel H. Kravitz; Maureen Durning; Thaddeus G. Golos

Collaboration


Dive into the Gennadiy I. Bondarenko's collaboration.

Top Co-Authors

Avatar

Thaddeus G. Golos

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Maureen Durning

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

Svetlana V. Dambaeva

Rosalind Franklin University of Medicine and Science

View shared research outputs
Top Co-Authors

Avatar

Richard L. Grendell

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

David W. Burleigh

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Heather A. Simmons

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Nancy G. Faith

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Andres Mejia

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Charles J. Czuprynski

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge