Network


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

Hotspot


Dive into the research topics where Michele L. Schotzko is active.

Publication


Featured researches published by Michele L. Schotzko.


Nature Communications | 2016

A rhesus macaque model of Asian-lineage Zika virus infection

Dawn M. Dudley; Matthew T. Aliota; Emma L. Mohr; Andrea M. Weiler; Gabrielle Lehrer-Brey; Kim L. Weisgrau; Mariel S. Mohns; Meghan E. Breitbach; Mustafa N. Rasheed; Christina M. Newman; Dane D. Gellerup; Louise H. Moncla; Jennifer Post; Nancy Schultz-Darken; Michele L. Schotzko; Jennifer M. Hayes; Josh Eudailey; M. Anthony Moody; Sallie R. Permar; Shelby L. O’Connor; Eva G. Rakasz; Heather A. Simmons; Saverio Capuano; Thaddeus G. Golos; Jorge E. Osorio; Thomas C. Friedrich; David H. O’Connor

Infection with Asian-lineage Zika virus (ZIKV) has been associated with Guillain–Barré syndrome and fetal abnormalities, but the underlying mechanisms remain poorly understood. Animal models of infection are thus urgently needed. Here we show that rhesus macaques are susceptible to infection by an Asian-lineage ZIKV closely related to strains currently circulating in the Americas. Following subcutaneous inoculation, ZIKV RNA is detected in plasma 1 day post infection (d.p.i.) in all animals (N=8, including 2 pregnant animals), and is also present in saliva, urine and cerebrospinal fluid. Non-pregnant and pregnant animals remain viremic for 21 days and for up to at least 57 days, respectively. Neutralizing antibodies are detected by 21 d.p.i. Rechallenge 10 weeks after the initial challenge results in no detectable virus replication, indicating protective immunity against homologous strains. Therefore, Asian-lineage ZIKV infection of rhesus macaques provides a relevant animal model for studying pathogenesis and evaluating potential interventions against human infection, including during pregnancy.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Rhesus monkey placental transgene expression after lentiviral gene transfer into preimplantation embryos

M. J. Wolfgang; Stephen G. Eisele; M. A. Browne; Michele L. Schotzko; M. A. Garthwaite; M. Durning; A. Ramezani; R. G. Hawley; James A. Thomson; Thaddeus G. Golos

Transgenic mice have provided invaluable information about gene function and regulation. However, because of marked differences between rodents and primates, some areas of human biology such as early embryonic development, aging, and maternal–fetal interactions would be best studied in a nonhuman primate model. Here, we report that gene transfer into rhesus monkey (Macaca mulatta) preimplantation embryos gives rise to transgenic placentas that express a reporter transgene (eGFP). Blastocysts resulting from culture of in vitro fertilized ova were transduced with a self-inactivating lentiviral vector and transferred into recipient females. One twin and one singleton pregnancy were produced from a single stimulation cycle, and one live rhesus monkey was born from each pregnancy. Placentas from all conceptuses showed expression of the transgene as detected by reverse transcription–PCR, ribonuclease protection assay, direct epifluorescence, immunohistochemistry, and Western blot analysis. Integration in somatic tissues of the offspring was not detected. A maternal immune response to the xenogeneic placental antigen was shown by the presence of anti-GFP antibodies in peripheral blood of the recipient females by day 99 of gestation (term = 165 days). These results demonstrate that transgene expression during gestation is compatible with successful pregnancy in nonhuman primates and provides an approach that could be broadly applicable to the development of novel models for primate biomedical research.


PLOS Pathogens | 2017

Highly efficient maternal-fetal Zika virus transmission in pregnant rhesus macaques

Sydney Nguyen; Kathleen M. Antony; Dawn M. Dudley; Sarah Kohn; Heather A. Simmons; Bryce Wolfe; M. Shahriar Salamat; Leandro B. C. Teixeira; Gregory J. Wiepz; Troy H. Thoong; Matthew T. Aliota; Andrea M. Weiler; Gabrielle L. Barry; Kim L. Weisgrau; Logan J. Vosler; Mariel S. Mohns; Meghan E. Breitbach; Laurel M. Stewart; Mustafa N. Rasheed; Christina M. Newman; Michael E. Graham; Oliver Wieben; Patrick A. Turski; Kevin M. Johnson; Jennifer Post; Jennifer M. Hayes; Nancy Schultz-Darken; Michele L. Schotzko; Josh Eudailey; Sallie R. Permar

Infection with Zika virus (ZIKV) is associated with human congenital fetal anomalies. To model fetal outcomes in nonhuman primates, we administered Asian-lineage ZIKV subcutaneously to four pregnant rhesus macaques. While non-pregnant animals in a previous study contemporary with the current report clear viremia within 10–12 days, maternal viremia was prolonged in 3 of 4 pregnancies. Fetal head growth velocity in the last month of gestation determined by ultrasound assessment of head circumference was decreased in comparison with biparietal diameter and femur length within each fetus, both within normal range. ZIKV RNA was detected in tissues from all four fetuses at term cesarean section. In all pregnancies, neutrophilic infiltration was present at the maternal-fetal interface (decidua, placenta, fetal membranes), in various fetal tissues, and in fetal retina, choroid, and optic nerve (first trimester infection only). Consistent vertical transmission in this primate model may provide a platform to assess risk factors and test therapeutic interventions for interruption of fetal infection. The results may also suggest that maternal-fetal ZIKV transmission in human pregnancy may be more frequent than currently appreciated.


Journal of Medical Primatology | 2001

Pregnancy and live birth from nonsurgical transfer of in vivo- and in vitro -produced blastocysts in the rhesus monkey

Michael J. Wolfgang; Stephen G. Eisele; Lisa Knowles; Melissa A. Browne; Michele L. Schotzko; Thaddeus G. Golos

Abstract: Embryo transfer in the rhesus monkey has been historically limited to transfer of cleavage stage embryos. In order to allow genetic manipulation of rhesus embryos in vitro, without using invasive surgical techniques, it is important to explore the transfer of morula and blastocyst stage embryos. Embryos were produced by in vitro fertilization from gonadotropin‐stimulated monkeys, or were obtained by nonsurgical uterine flushing of naturally mated or artificially inseminated females. Nonsurgical transfer was accomplished by inserting a metal guide through the cervix into the uterus, after which a hollow cell sampler was inserted over the guide. The guide was removed and a catheter was inserted containing one to five embryos. Several pregnancies resulted from in vitro‐ and in vivo ‐derived blastocysts, and two pregnancies were carried to term resulting in one live birth. Blood samples were collected regularly to monitor plasma levels of chorionic gonadotropin, luteinizing hormone, and progesterone. The recipients received progesterone as a subcutaneous implant or daily injections from the day of transfer. The approach described in this study provides the opportunity to explore transgenic and chimeric models in the monkey by the development of noninvasive methods to transfer late‐stage embryos that have been manipulated in vitro.


PLOS ONE | 2018

Ocular and uteroplacental pathology in a macaque pregnancy with congenital Zika virus infection.

Emma L. Mohr; Lindsey N. Block; Christina M. Newman; Laurel M. Stewart; Michelle R. Koenig; Matthew Semler; Meghan E. Breitbach; Leandro B. C. Teixeira; Xiankun Zeng; Andrea M. Weiler; Gabrielle L. Barry; Troy H. Thoong; Gregory J. Wiepz; Dawn M. Dudley; Heather A. Simmons; Andres Mejia; Terry K. Morgan; M. Shahriar Salamat; Sarah Kohn; Kathleen M. Antony; Matthew T. Aliota; Mariel S. Mohns; Jennifer M. Hayes; Nancy Schultz-Darken; Michele L. Schotzko; Eric Peterson; Saverio Capuano; Jorge E. Osorio; Shelby L. O’Connor; Thomas C. Friedrich

Congenital Zika virus (ZIKV) infection impacts fetal development and pregnancy outcomes. We infected a pregnant rhesus macaque with a Puerto Rican ZIKV isolate in the first trimester. The pregnancy was complicated by preterm premature rupture of membranes (PPROM), intraamniotic bacterial infection and fetal demise 49 days post infection (gestational day 95). Significant pathology at the maternal-fetal interface included acute chorioamnionitis, placental infarcts, and leukocytoclastic vasculitis of the myometrial radial arteries. ZIKV RNA was disseminated throughout fetal tissues and maternal immune system tissues at necropsy, as assessed by quantitative RT-PCR for viral RNA. Replicating ZIKV was identified in fetal tissues, maternal uterus, and maternal spleen by fluorescent in situ hybridization for viral replication intermediates. Fetal ocular pathology included a choroidal coloboma, suspected anterior segment dysgenesis, and a dysplastic retina. This is the first report of ocular pathology and prolonged viral replication in both maternal and fetal tissues following congenital ZIKV infection in a rhesus macaque. PPROM followed by fetal demise and severe pathology of the visual system have not been described in macaque congenital ZIKV infection previously. While this case of ZIKV infection during pregnancy was complicated by bacterial infection with PPROM, the role of ZIKV on this outcome cannot be precisely defined, and further nonhuman primate studies will determine if increased risk for PPROM or other adverse pregnancy outcomes are associated with congenital ZIKV infection.


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.


PLOS Pathogens | 2018

Molecularly barcoded Zika virus libraries to probe in vivo evolutionary dynamics

Matthew T. Aliota; Dawn M. Dudley; Christina M. Newman; James Weger-Lucarelli; Laurel M. Stewart; Michelle R. Koenig; Meghan E. Breitbach; Andrea M. Weiler; Matthew Semler; Gabrielle L. Barry; Katie R. Zarbock; Amelia K. Haj; Ryan V. Moriarty; Mariel S. Mohns; Emma L. Mohr; Vanessa Venturi; Nancy Schultz-Darken; Eric Peterson; Wendy Newton; Michele L. Schotzko; Heather A. Simmons; Andres Mejia; Jennifer M. Hayes; Saverio Capuano; Miles P. Davenport; Thomas C. Friedrich; Gregory D. Ebel; Shelby L. O’Connor; David H. O’Connor

Defining the complex dynamics of Zika virus (ZIKV) infection in pregnancy and during transmission between vertebrate hosts and mosquito vectors is critical for a thorough understanding of viral transmission, pathogenesis, immune evasion, and potential reservoir establishment. Within-host viral diversity in ZIKV infection is low, which makes it difficult to evaluate infection dynamics. To overcome this biological hurdle, we constructed a molecularly barcoded ZIKV. This virus stock consists of a “synthetic swarm” whose members are genetically identical except for a run of eight consecutive degenerate codons, which creates approximately 64,000 theoretical nucleotide combinations that all encode the same amino acids. Deep sequencing this region of the ZIKV genome enables counting of individual barcodes to quantify the number and relative proportions of viral lineages present within a host. Here we used these molecularly barcoded ZIKV variants to study the dynamics of ZIKV infection in pregnant and non-pregnant macaques as well as during mosquito infection/transmission. The barcoded virus had no discernible fitness defects in vivo, and the proportions of individual barcoded virus templates remained stable throughout the duration of acute plasma viremia. ZIKV RNA also was detected in maternal plasma from a pregnant animal infected with barcoded virus for 67 days. The complexity of the virus population declined precipitously 8 days following infection of the dam, consistent with the timing of typical resolution of ZIKV in non-pregnant macaques and remained low for the subsequent duration of viremia. Our approach showed that synthetic swarm viruses can be used to probe the composition of ZIKV populations over time in vivo to understand vertical transmission, persistent reservoirs, bottlenecks, and evolutionary dynamics.


bioRxiv | 2017

Ocular and uteroplacental pathology in macaque congenital Zika virus infection

Emma L. Mohr; Lindsey N. Block; Christina M. Newman; Laurel M. Stewart; Michelle R. Koenig; Matthew Semler; Meghan E. Breitbach; Leandro B. C. Teixeira; Xiankun Zeng; Andrea M. Weiler; Gabrielle L. Barry; Troy H. Thoong; Gregory J. Wiepz; Dawn M. Dudley; Heather A. Simmons; Andres Mejia; Terry K. Morgan; M. Shahriar Salamat; Sarah Kohn; Kathleen M. Antony; Matthew T. Aliota; Mariel S. Mohns; Jennifer M. Hayes; Nancy Schultz-Darken; Michele L. Schotzko; Eric Peterson; Saverio Capuano; Jorge E. Osorio; Shelby L. O'Connor; Thomas C. Friedrich

Congenital Zika virus (ZIKV) infection impacts fetal development and pregnancy outcomes. We infected a pregnant rhesus macaque with a Puerto Rican ZIKV isolate in the first trimester. The pregnancy was complicated by preterm premature rupture of membranes (PPROM) and fetal demise 49 days post infection (gestational day 95). Significant pathology at the maternal-fetal interface included acute chorioamnionitis, placental infarcts, and leukocytoclastic vasculitis of the myometrial radial arteries. ZIKV RNA was disseminated throughout the fetus tissues and maternal immune system at necropsy, as assessed by quantitative RT-PCR for viral RNA. Replicating ZIKV was identified in fetal tissues, maternal lymph node, and maternal spleen by fluorescent in situ hybridization for viral replication intermediates. Fetal ocular pathology included a choroidal coloboma, suspected anterior segment dysgenesis, and a dysplastic retina. This is the first report of ocular pathology and prolonged viral replication in both maternal and fetal tissues following congenital ZIKV infection in rhesus macaques. PPROM followed by fetal demise and severe pathology of the visual system have not been described in macaque congenital infection previously; further nonhuman primate studies are needed to determine if an increased risk for PPROM is associated with congenital Zika virus infection. Author summary A ZIKV infection during pregnancy is associated with malformations in fetal development including, but not limited to, ocular and brain anomalies, such as microcephaly, and stillbirth. The development of an accurate pregnancy model to study the effects of ZIKV will provide insight into vertical transmission, ZIKV tissue distribution, and fetal injury and malformations. Non-human primates closely resemble human in terms of the reproductive system, immunity, placentation and pregnancy. Our study demonstrates that the rhesus macaque is a compelling model in which to study ZIKV during pregnancy due to similar outcomes between the human and rhesus macaque. These similarities include prolonged viremia, vertical transmission, adverse pregnancy outcomes and fetal pathology, including defects in the visual system.


Molecular Reproduction and Development | 2002

Efficient method for expressing transgenes in nonhuman primate embryos using a stable episomal vector

Michael J. Wolfgang; Vivienne S. Marshall; Stephen G. Eisele; Michele L. Schotzko; James A. Thomson; Thaddeus G. Golos


Placenta | 2017

Vertical Transmission of Zika Virus (ZIKV) in Rhesus Macaques

Lindsey N. Block; Leandro B. C. Teixeira; Xiankun Zeng; Dawn M. Dudley; Kathleen M. Antony; Heather A. Simmons; Gregory J. Wiepz; Matthew T. Aliota; Gabrielle L. Barry; Mariel S. Mohns; Meghan E. Breitbach; Laurel M. Stewart; Michele L. Schotzko; Sallie R. Permar; Emma L. Mohr; Saverio Capuano; Jorge E. Osorio; Thomas C. Friedrich; David H. O'Connor; Thaddeus G. Golos

Collaboration


Dive into the Michele L. Schotzko's collaboration.

Top Co-Authors

Avatar

Heather A. Simmons

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Thaddeus G. Golos

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Dawn M. Dudley

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Mariel S. Mohns

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Matthew T. Aliota

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Meghan E. Breitbach

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Andrea M. Weiler

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

Christina M. Newman

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Emma L. Mohr

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge