Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
eNeuro ; 10(11)2023 Nov.
Article in English | MEDLINE | ID: mdl-37957008

ABSTRACT

Because of the legalization of Cannabis in many jurisdictions and the trend of increasing Δ9-tetrahydrocannabinol (THC) content in Cannabis products, an urgent need exists to understand the impact of Cannabis use during pregnancy on fetal neurodevelopment and behavior. To this end, we exposed female Sprague Dawley rats to Cannabis smoke daily from gestational day 6 to 20 or room air. Maternal reproductive parameters, offspring behavior, and gene expression in the offspring amygdala were assessed. Body temperature was decreased in dams following smoke exposure and more fecal boli were observed in the chambers before and after smoke exposure in dams exposed to smoke. Maternal weight gain, food intake, gestational length, litter number, and litter weight were not altered by exposure to Cannabis smoke. A significant increase in the male-to-female ratio was noted in the Cannabis-exposed litters. In adulthood, male and female Cannabis smoke-exposed offspring explored the inner zone of an open field significantly less than control offspring. Gestational Cannabis smoke exposure did not affect behavior on the elevated plus maze test or social interaction test in the offspring. Cannabis offspring were better at visual pairwise discrimination and reversal learning tasks conducted in touchscreen-equipped operant conditioning chambers. Analysis of gene expression in the adult amygdala using RNA sequencing revealed subtle changes in genes related to development, cellular function, and nervous system disease in a subset of the male offspring. These results demonstrate that repeated exposure to high-THC Cannabis smoke during gestation alters maternal physiological parameters, sex ratio, and anxiety-like behaviors in the adulthood offspring.


Subject(s)
Cannabis , Prenatal Exposure Delayed Effects , Pregnancy , Rats , Male , Female , Animals , Humans , Rats, Sprague-Dawley , Sex Ratio , Reproduction , Gene Expression
2.
Emerg Microbes Infect ; 12(1): 2147021, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36369716

ABSTRACT

Congenital virus infections, for example cytomegalovirus and rubella virus infections, commonly affect the central nervous and hematological systems in fetuses and offspring. However, interactions between emerging congenital Zika virus and hematological system-bone marrow and blood-in fetuses and offspring are mainly unknown. Our overall goal was to determine whether silent in utero Zika virus infection can cause functional and molecular footprints in the bone marrow and blood of fetuses and offspring. We specifically focused on silent fetal infection because delayed health complications in initially asymptomatic offspring were previously demonstrated in animal and human studies. Using a well-established porcine model for Zika virus infection and a set of cellular and molecular experimental tools, we showed that silent in utero infection causes multi-organ inflammation in fetuses and local inflammation in the fetal bone marrow. In utero infection also caused footprints in the offspring bone marrow and PBMCs. These findings should be considered in a broader clinical context because of growing concerns about health sequelae in cohorts of children affected with congenital Zika virus infection in the Americas. Understanding virus-induced molecular mechanisms of immune activation and inflammation in fetuses may provide targets for early in utero interventions. Also, identifying early biomarkers of in utero-acquired immunopathology in offspring may help to alleviate long-term sequelae.


Subject(s)
Zika Virus Infection , Zika Virus , Child , Female , Humans , Animals , Swine , Zika Virus/genetics , Bone Marrow , Blood Cells/pathology , Inflammation
3.
Front Immunol ; 13: 943481, 2022.
Article in English | MEDLINE | ID: mdl-35983032

ABSTRACT

Increasing the number of CpG dinucleotides in RNA viral genomes, while preserving the original amino acid composition, leads to impaired infection which does not cause disease. Beneficially, impaired infection evokes antiviral host immune responses providing a cutting-edge vaccine approach. For example, we previously showed that CpG-enriched Zika virus variants cause attenuated infection phenotypes and protect against lethal challenge in mice. While CpG recoding is an emerging and promising vaccine approach, little is known about infection phenotypes caused by recoded viruses in vivo, particularly in non-rodent species. Here, we used well-established mouse and porcine models to study infection phenotypes of the CpG-enriched neurotropic and congenital virus-Zika virus, directly in the target tissues-the brain and placenta. Specifically, we used the uttermost challenge and directly injected mice intracerebrally to compare infection phenotypes caused by wild-type and two CpG-recoded Zika variants and model the scenario where vaccine strains breach the blood-brain barrier. Also, we directly injected porcine fetuses to compare in utero infection phenotypes and model the scenario where recoded vaccine strains breach the placental barrier. While overall infection kinetics were comparable between wild-type and recoded virus variants, we found convergent phenotypical differences characterized by reduced pathology in the mouse brain and reduced replication of CpG-enriched variants in fetal lymph nodes. Next, using next-generation sequencing for the whole virus genome, we compared the stability of de novo introduced CpG dinucleotides during prolonged virus infection in the brain and placenta. Most de novo introduced CpG dinucleotides were preserved in sequences of recoded Zika viruses showing the stability of vaccine variants. Altogether, our study emphasized further directions to fine-tune the CpG recoding vaccine approach for better safety and can inform future immunization strategies.


Subject(s)
Viruses , Zika Virus Infection , Zika Virus , Animals , Brain , Female , Fetus , Genome, Viral , Lymph Nodes , Mice , Phenotype , Placenta , Pregnancy , Swine , Viruses/genetics
4.
Methods Mol Biol ; 2410: 289-302, 2022.
Article in English | MEDLINE | ID: mdl-34914053

ABSTRACT

Experimental increase of cytosine-phosphate-guanine (CpG) dinucleotides in an RNA virus genome impairs infection. Beneficially, this weak infection may lead to robust antiviral host immunity providing a cutting-edge approach for vaccines. For example, we have recently demonstrated that recoded Zika virus variants with the increased CpG content showed considerable attenuated infection phenotypes and protection against lethal challenge in mice. Here, we describe the workflow for the design and generation of CpG-recoded Zika virus vaccine candidates. The workflow can be adapted for other viruses.


Subject(s)
Zika Virus Infection , Zika Virus , Animals , Antibodies, Viral , Mice , Phosphates , Viral Vaccines , Zika Virus/immunology , Zika Virus Infection/prevention & control
5.
Viruses ; 13(9)2021 09 14.
Article in English | MEDLINE | ID: mdl-34578408

ABSTRACT

The host's immune status may affect virus evolution. Little is known about how developing fetal and placental immune milieus affect virus heterogeneity. This knowledge will help us better understand intra-host virus evolution and how new virus variants emerge. The goal of our study was to find out whether the isolated in utero environment-an environment with specialized placental immunity and developing fetal immunity-supports the emergence of RNA and DNA virus variants. We used well-established porcine models for isolated Zika virus (RNA virus) and porcine circovirus 2 (DNA virus) fetal infections. We found that the isolated in utero environment was conducive to the emergence of RNA and DNA virus variants. Next-generation sequencing of nearly whole virus genomes and validated bioinformatics pipelines identified both unique and convergent single nucleotide variations in virus genomes isolated from different fetuses. Zika virus and PCV2 in utero evolution also resulted in single nucleotide variations previously reported in the human and porcine field samples. These findings should encourage further studies on virus evolution in placenta and fetuses, to better understand how virus variants emerge and how in utero viral evolution affects congenital virus transmission and pathogenicity.


Subject(s)
Circovirus/genetics , Directed Molecular Evolution , Genetic Variation , Placenta/virology , Uterus/virology , Zika Virus/genetics , Animals , Cellular Microenvironment , Chlorocebus aethiops , Circovirus/physiology , Female , Fetus/virology , Genetic Heterogeneity , High-Throughput Nucleotide Sequencing , Placenta/immunology , Pregnancy , Swine , Uterus/immunology , Vero Cells , Viral Load , Zika Virus/physiology
6.
Methods Mol Biol ; 2142: 181-195, 2020.
Article in English | MEDLINE | ID: mdl-32367368

ABSTRACT

Pigs are highly relevant to model human in utero Zika virus (ZIKV) infection because both species have similar physiology, genetics, immunity, fetal brain development, and postnatal brain growth. The virus causes persistent in utero infection and replicates in the fetal brain, fetal membranes, and placenta. Subclinical persistent in utero infection in mid-gestation also increases interferon alpha (IFN-α) levels in fetal blood plasma and amniotic fluid. Moreover, we demonstrated altered IFN-α responses in porcine offspring affected with subclinical in utero ZIKV infection. Elevated levels of in utero type I interferons were suggested to play a role in fetal pathology. Thus, the porcine model may provide an understanding of ZIKV-induced immunopathology in fetuses and sequelae in offspring, which is important for the development of targeted interventions. Here, we describe surgery, ultrasound-guided in utero injection, postoperative monitoring, sampling, and cytokine testing protocols.


Subject(s)
Disease Models, Animal , Fetal Diseases , Interferon-alpha/metabolism , Pregnancy Complications, Infectious , Swine , Zika Virus Infection , Amniotic Fluid/metabolism , Animals , Blood Chemical Analysis/methods , Blood Chemical Analysis/veterinary , Female , Fetal Blood/metabolism , Fetal Diseases/diagnostic imaging , Fetal Diseases/metabolism , Fetal Diseases/pathology , Fetal Diseases/virology , Fetoscopy/methods , Fetoscopy/veterinary , Injections , Interferon-alpha/analysis , Interferon-alpha/blood , Pregnancy , Pregnancy Complications, Infectious/diagnostic imaging , Pregnancy Complications, Infectious/metabolism , Pregnancy Complications, Infectious/pathology , Ultrasonography, Interventional/methods , Ultrasonography, Interventional/veterinary , Ultrasonography, Prenatal/methods , Ultrasonography, Prenatal/veterinary , Zika Virus/physiology , Zika Virus Infection/diagnostic imaging , Zika Virus Infection/metabolism , Zika Virus Infection/pathology
7.
PLoS Pathog ; 15(11): e1008038, 2019 11.
Article in English | MEDLINE | ID: mdl-31725819

ABSTRACT

Zika virus (ZIKV) infection during human pregnancy may lead to severe fetal pathology and debilitating impairments in offspring. However, the majority of infections are subclinical and not associated with evident birth defects. Potentially detrimental life-long health outcomes in asymptomatic offspring evoke high concerns. Thus, animal models addressing sequelae in offspring may provide valuable information. To induce subclinical infection, we inoculated selected porcine fetuses at the mid-stage of development. Inoculation resulted in trans-fetal virus spread and persistent infection in the placenta and fetal membranes for two months. Offspring did not show congenital Zika syndrome (e.g., microcephaly, brain calcifications, congenital clubfoot, arthrogryposis, seizures) or other visible birth defects. However, a month after birth, a portion of offspring exhibited excessive interferon alpha (IFN-α) levels in blood plasma in a regular environment. Most affected offspring also showed dramatic IFN-α shutdown during social stress providing the first evidence for the cumulative impact of prenatal ZIKV exposure and postnatal environmental insult. Other eleven cytokines tested before and after stress were not altered suggesting the specific IFN-α pathology. While brains from offspring did not have histopathology, lesions, and ZIKV, the whole genome expression analysis of the prefrontal cortex revealed profound sex-specific transcriptional changes that most probably was the result of subclinical in utero infection. RNA-seq analysis in the placenta persistently infected with ZIKV provided independent support for the sex-specific pattern of in utero-acquired transcriptional responses. Collectively, our results provide strong evidence that two hallmarks of fetal ZIKV infection, altered type I IFN response and molecular brain pathology can persist after birth in offspring in the absence of congenital Zika syndrome.


Subject(s)
Brain/pathology , Fetal Diseases/epidemiology , Fetus/virology , Interferon-alpha/metabolism , Pregnancy Complications, Infectious/epidemiology , Uterus/virology , Zika Virus Infection/virology , Animals , Antiviral Agents/metabolism , Brain/metabolism , Brain/virology , Communicable Diseases/transmission , Communicable Diseases/virology , Female , Fetal Diseases/metabolism , Fetal Diseases/virology , Fetus/metabolism , Fetus/pathology , Male , Pregnancy , Pregnancy Complications, Infectious/metabolism , Pregnancy Complications, Infectious/virology , Sex Factors , Swine , Uterus/metabolism , Uterus/pathology , Zika Virus/pathogenicity , Zika Virus Infection/pathology , Zika Virus Infection/transmission , Zika Virus Infection/veterinary
8.
Emerg Microbes Infect ; 8(1): 1098-1107, 2019.
Article in English | MEDLINE | ID: mdl-31340725

ABSTRACT

Studies in mice showed that African Zika virus (ZIKV) strains cause more damage in embryos. These studies, however, were limited to the mouse-adapted African MR766 strain or infection at early gestation. Here, we compared infection of Asian and African strains in the fetal pig model at midgestation. Both strains caused fetal infection. ZIKV was detected in placenta, amniotic membrane, amniotic fluid, fetal blood, and brain. The African strain produced more vigorous in utero infection as represented by more efficient virus transmission between siblings, and higher viral loads in fetal organs and membranes. Infection with both strains was associated with reduced fetal brain weight and increased number of placental CD163-positive cells, as well as elevated in utero interferon alpha and cortisol levels. This is the first large animal model study which demonstrated that African strain of ZIKV, with no passage history in experimental animals, can cause persistent infection in fetuses and fetal membranes at midgestation. Our studies also suggest that similar to Asian strains, ZIKV of African lineage might cause silent pathology which is difficult to identify in deceptively healthy fetuses. The findings emphasize the need for further studies to highlight the impact of ZIKV heterogeneity on infection outcomes during pregnancy.


Subject(s)
Fetal Diseases/virology , Pregnancy Complications, Infectious/virology , Zika Virus Infection/virology , Zika Virus/physiology , Animals , Brain/virology , Disease Models, Animal , Female , Humans , Placenta/virology , Pregnancy , Swine , Uterus/virology , Zika Virus/classification , Zika Virus/genetics , Zika Virus Infection/transmission
9.
Front Immunol ; 10: 3077, 2019.
Article in English | MEDLINE | ID: mdl-32038625

ABSTRACT

Experimental increase of CpG dinucleotides in an RNA virus genome impairs infection providing a promising approach for vaccine development. While CpG recoding is an emerging and promising vaccine approach, little is known about infection phenotypes caused by recoded viruses in vivo. For example, infection phenotypes, immunogenicity, and protective efficacy induced by CpG-recoded viruses in different age groups were not studied yet. This is important, because attenuation of infection phenotypes caused by recoded viruses may depend on the population-based expression of cellular components targeting viral CpG dinucleotides. In the present study, we generated several Zika virus (ZIKV) variants with the increasing CpG content and compared infection in neonatal and adult mice. Increasing the CpG content caused host-age-dependent attenuation of infection with considerable attenuation in neonates and high attenuation in adults. Expression of the zinc-finger antiviral protein (ZAP)-the host protein targeting viral CpG dinucleotides-was also age-dependent. Similar to the wild-type virus, ZIKV variants with the increased CpG content evoked robust cellular and humoral immune responses and protection against lethal challenge. Collectively, the host age should be accounted for in future studies on mechanisms targeting viral CpG dinucleotides, development of safe dinucleotide recoding strategies, and applications of CpG-recoded vaccines.


Subject(s)
CpG Islands/genetics , Genome, Viral/genetics , Viral Vaccines/genetics , Zika Virus Infection/immunology , Zika Virus/physiology , Age Factors , Animals , Animals, Newborn , Cell Line , Disease Resistance , Humans , Immunity, Cellular , Immunity, Heterologous , Immunity, Humoral , Mice , Mice, Inbred C57BL , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...