Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
International Journal of Pediatrics-Mashhad ; 11(4):17558-17571, 2023.
Article in English | Web of Science | ID: covidwho-2328100

ABSTRACT

Background: Viral infection during pregnancy induces pro-inflammatory cytokines and increases the level of IL-6, which as a measure of the inflammation of the mother's immune system is effective on the development of the brain of the fetus and increases the rate of mental disorders of the fetus. The possible effect of the mother's COVID-19 on children, if any, is still unknown. Therefore, this study was conducted with the aim of investigating and comparing the consequences of neurodevelopment and growth at the age of 6 months in infants among mothers with/without exposure to COVID-19 during pregnancy in Birjand, 2021.Methods: A total of 161 pregnant women with PCR-positive coronavirus and 181 pregnant women without infection were selected by simple random sampling. Demographic characteristics including age, occupation, education, gender, nationality, sex of babies, along with data related to the neurodevelopmental status of infants by the use of ASQ questionnaire were extracted and entered into SPSS 16 software. For descriptive statistics, Mean, standard deviation, and frequency distribution tables were used, and inferential analyses were conducted by the use of Chi-square, t-test, and analysis of variance at a significance level of less than 0.05. The disturbance was equal / lower than 1 SD in each of ASQ-3 score domains.Results: In this cohort study, demographic characteristics in the exposed and unexposed groups were not significantly different. There was no statistically significant difference between the average score of the ASQ_3 questionnaires as well as the average score in the areas of communication, gross motor, fine motor, problem-solving, and social personality among infants born from mothers with and without COVID-19. Also, there was no significant difference in the incidence of disorders in different fields of neurodevelopment, except for the field of establishing communication between the two groups of exposed and unexposed. On the other hand, cesarean delivery was significantly higher in pregnant mothers with COVID-19 during pregnancy. However, there was no significant relationship between the incidence of COVID-19 and the rate of premature delivery, and low birth weight neonates. In addition, the severity and time of the corona infection during pregnancy did not have any significant effect on the neurodevelopment of infants with intrauterine exposure to COVID-19.Conclusion: According to the results of the study, infection with COVID-19 does not increase the incidence of developmental disorders in 6-month-old infants. Furthermore, it does not increase low birth weight and premature delivery. Due to the limitations of the study, it is suggested to conduct a study with a larger number of pregnant mothers with a severe form of the disease, especially in the first trimester of pregnancy, and to follow up with the babies at an older age.

2.
Int J Mol Sci ; 24(6)2023 Mar 13.
Article in English | MEDLINE | ID: covidwho-2284960

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopmental disorder (NDD) characterized by impairments in social communication, repetitive behaviors, restricted interests, and hyperesthesia/hypesthesia caused by genetic and/or environmental factors. In recent years, inflammation and oxidative stress have been implicated in the pathogenesis of ASD. In this review, we discuss the inflammation and oxidative stress in the pathophysiology of ASD, particularly focusing on maternal immune activation (MIA). MIA is a one of the common environmental risk factors for the onset of ASD during pregnancy. It induces an immune reaction in the pregnant mother's body, resulting in further inflammation and oxidative stress in the placenta and fetal brain. These negative factors cause neurodevelopmental impairments in the developing fetal brain and subsequently cause behavioral symptoms in the offspring. In addition, we also discuss the effects of anti-inflammatory drugs and antioxidants in basic studies on animals and clinical studies of ASD. Our review provides the latest findings and new insights into the involvements of inflammation and oxidative stress in the pathogenesis of ASD.


Subject(s)
Autism Spectrum Disorder , Prenatal Exposure Delayed Effects , Humans , Pregnancy , Animals , Female , Autism Spectrum Disorder/pathology , Neuroinflammatory Diseases , Inflammation/complications , Oxidative Stress
3.
Cells ; 12(5)2023 02 25.
Article in English | MEDLINE | ID: covidwho-2281172

ABSTRACT

The developmental origins of health and disease (DOHaD) indicate that fetal tissues and organs in critical and sensitive periods of development are susceptible to structural and functional changes due to the adverse environment in utero. Maternal immune activation (MIA) is one of the phenomena in DOHaD. Exposure to maternal immune activation is a risk factor for neurodevelopmental disorders, psychosis, cardiovascular diseases, metabolic diseases, and human immune disorders. It has been associated with increased levels of proinflammatory cytokines transferred from mother to fetus in the prenatal period. Abnormal immunity induced by MIA includes immune overreaction or immune response failure in offspring. Immune overreaction is a hypersensitivity response of the immune system to pathogens or allergic factor. Immune response failure could not properly fight off various pathogens. The clinical features in offspring depend on the gestation period, inflammatory magnitude, inflammatory type of MIA in the prenatal period, and exposure to prenatal inflammatory stimulation, which might induce epigenetic modifications in the immune system. An analysis of epigenetic modifications caused by adverse intrauterine environments might allow clinicians to predict the onset of diseases and disorders before or after birth.


Subject(s)
Prenatal Exposure Delayed Effects , Pregnancy , Female , Humans , Immune System/metabolism , Parturition , Cytokines , Mothers
4.
Phenotyping of Human iPSC-derived Neurons: Patient-Driven Research ; : 173-200, 2022.
Article in English | Scopus | ID: covidwho-2149084

ABSTRACT

The developing fetus, while generally safe from the outside world, is often exposed to any one of a number of toxins, drugs, infectious microbes, and maternal antibodies and cytokines that adversely affect the developing brain. This can have life-long consequences on cognitive function and behavior. Cerebral organoids derived from induced pluripotent stem cells (iPSCs) are used as a model system to study the first trimester brain, providing researchers with an opportunity to identify underlying molecular pathways that are disrupted by potentially dangerous environmental exposures. In addition, using patient-specific iPSCs allows researchers to study gene x environment interactions. Such studies could lead to the development of novel therapies for at-risk fetuses. © 2023 Elsevier Inc. All rights reserved.

5.
Front Neurosci ; 16: 943903, 2022.
Article in English | MEDLINE | ID: covidwho-1917227

ABSTRACT

[This corrects the article DOI: 10.3389/fnins.2022.834058.].

6.
Behav Brain Res ; 430: 113930, 2022 07 26.
Article in English | MEDLINE | ID: covidwho-1850692

ABSTRACT

Evidence suggests that early life adversity, such as maternal immune activation (MIA), can alter brain development in the offspring and confer increased risk for psychopathology and psychiatric illness in later life. In this study, the long-term effects of MIA, post-weaning social isolation, and the combination were assessed on behavioural and immunological profiles in adult male and female offspring. On gestation day 12.5, pregnant mice were weighed and injected with either polyinosinic:polycytidylic acid (5 mg/kg) or saline and cytokines levels were assayed 3 hrs later to confirm immune activation. The behaviour and immunological profiles of male and female offspring were examined in adolescence (P34-36), and adulthood (P55-80). MIA induced an increase in the pro-inflammatory cytokine IL-6 in pregnant dams three hours after administration (p < 0.001) that correlated with a decrease in body temperature (p < 0.05). The effect of MIA on the immunological phenotype of the offspring was evident in adolescence, but not in adulthood. MIA selectively induced hypoactivity in adolescent males, a phenotype that persisted until adulthood, but had no effect on cognition in males or females. In contrast, social isolation stress from adolescence resulted in impaired sociability (p < 0.05) and increased anxiety (p < 0.05) particularly in adult females. There was no synergistic effect of the dual-hit on immune parameters, sociability, anxiety or cognitive behaviours. Given the negative impact and sex-dependent effects of SI stress on locomotor and anxiety-like behaviour, future investigations should examine whether the health risks of social isolation, such as that experience during the COVID-19 pandemic, are mediated through increased anxiety.


Subject(s)
COVID-19 , Prenatal Exposure Delayed Effects , Schizophrenia , Adolescent , Adult , Animals , Behavior, Animal/physiology , Cytokines/pharmacology , Disease Models, Animal , Endophenotypes , Female , Humans , Male , Mice , Pandemics , Poly I-C/pharmacology , Pregnancy , Social Isolation , Weaning
7.
Brain Behav Immun ; 103: 63-72, 2022 07.
Article in English | MEDLINE | ID: covidwho-1767916

ABSTRACT

IMPORTANCE: Animal studies show that Maternal Immune Activation (MIA) may have detrimental effects on fetal brain development. Clinical studies provide evidence for structural brain abnormalities in human neonates following MIA, but no study has investigated the long-term effects of MIA (as measured with biomarkers) on human brain morphology ten years after the exposure. OBJECTIVE: Our aim was to evaluate the long-term impact of MIA on brain morphology in 10-year-old children, including the possible mediating role of gestational age at birth. DESIGN: We leveraged data from Generation R, a large-scale prospective pregnancy cohort study. Pregnant women were included between 2002 and 2006, and their children were invited to participate in the MRI study between 2013 and 2015. To be included, mother-child dyads had to have data on maternal C-reactive protein levels during gestation and a good quality MRI-scan of the child's brain at age 10 years. Of the 3,992 children scanned, a total of 2,053 10-year-old children were included in this study. EXPOSURE: Maternal C-reactive protein was measured in the first 18 weeks of gestation. For the analyses we used both a continuous approach as well as a categorical approach based on clinical cut-offs to determine if there was a dose-response relationship. MAIN OUTCOMES AND MEASURES: High-resolution MRI brain morphology measures were used as the primary outcome. Gestational age at birth, established using ultrasound, was included as a mediator using a causal mediation analysis. Corrections were made for relevant confounders and multiple comparisons. Biological sex was investigated as moderator. RESULTS: We found a direct association between continuous MIA and lower cerebellar volume. In girls, we demonstrated a negative indirect association between continuous MIA and total brain volume, through the mediator gestational age at birth. We observed no associations with categorical MIA after multiple testing correction. CONCLUSION AND RELEVANCE: Our results suggest sex-specific long-term effects in brain morphology after MIA. Categorical analyses suggest that this association might be driven by acute infections or other sources of severe inflammation, which is of clinical relevance given that the COVID-19 pandemic is currently affecting millions of pregnant women worldwide.


Subject(s)
COVID-19 , Prenatal Exposure Delayed Effects , Animals , Brain/diagnostic imaging , C-Reactive Protein , Child , Cohort Studies , Female , Humans , Male , Pandemics , Pregnancy , Prospective Studies
8.
Front Psychiatry ; 13: 823096, 2022.
Article in English | MEDLINE | ID: covidwho-1731853

ABSTRACT

Autism spectrum disorder (ASD) is the commonest neurodevelopmental disability. It is a highly complex disorder with an increasing prevalence and an unclear etiology. Consensus indicates that ASD arises as a genetically modulated, and environmentally influenced condition. Although pathogenic rare genetic variants are detected in around 20% of cases of ASD, no single factor is responsible for the vast majority of ASD cases or that explains their characteristic clinical heterogeneity. However, a growing body of evidence suggests that ASD susceptibility involves an interplay between genetic factors and environmental exposures. One such environmental exposure which has received significant attention in this regard is maternal immune activation (MIA) resulting from bacterial or viral infection during pregnancy. Reproducible rodent models of ASD are well-established whereby induction of MIA in pregnant dams, leads to offspring displaying neuroanatomical, functional, and behavioral changes analogous to those seen in ASD. Blockade of specific inflammatory cytokines such as interleukin-17A during gestation remediates many of these observed behavioral effects, suggesting a causative or contributory role. Here, we review the growing body of animal and human-based evidence indicating that interleukin-17A may mediate the observed effects of MIA on neurodevelopmental outcomes in the offspring. This is particularly important given the current corona virus disease-2019 (COVID-19) pandemic as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection during pregnancy is a potent stimulator of the maternal immune response, however the long-term effects of maternal SARS-CoV-2 infection on neurodevelopmental outcomes is unclear. This underscores the importance of monitoring neurodevelopmental outcomes in children exposed to SARS-CoV-2-induced MIA during gestation.

9.
Biol Psychiatry Cogn Neurosci Neuroimaging ; 7(5): 510-523, 2022 05.
Article in English | MEDLINE | ID: covidwho-1729590

ABSTRACT

Pregnant women represent a uniquely vulnerable population during an infectious disease outbreak, such as the COVID-19 pandemic. Although we are at the early stages of understanding the specific impact of SARS-CoV-2 exposure during pregnancy, mounting epidemiological evidence strongly supports a link between exposure to a variety of maternal infections and an increased risk for offspring neurodevelopmental disorders. Inflammatory biomarkers identified from archived or prospectively collected maternal biospecimens suggest that the maternal immune response is the critical link between infection during pregnancy and altered offspring neurodevelopment. This maternal immune activation (MIA) hypothesis has been tested in animal models by artificially activating the immune system during pregnancy and evaluating the neurodevelopmental consequences in MIA-exposed offspring. Although the vast majority of MIA model research is carried out in rodents, the nonhuman primate model has emerged in recent years as an important translational tool. In this review, we briefly summarize human epidemiological studies that have prompted the development of translationally relevant MIA models. We then highlight notable similarities between humans and nonhuman primates, including placental structure, pregnancy physiology, gestational timelines, and offspring neurodevelopmental stages, that provide an opportunity to explore the MIA hypothesis in species more closely related to humans. Finally, we provide a comprehensive review of neurodevelopmental alterations reported in current nonhuman primate models of maternal infection and discuss future directions for this promising area of research.


Subject(s)
COVID-19 , Neurodevelopmental Disorders , Prenatal Exposure Delayed Effects , Animals , Disease Models, Animal , Female , Humans , Macaca mulatta , Neurodevelopmental Disorders/etiology , Pandemics , Placenta , Pregnancy , SARS-CoV-2
11.
Brain Behav Immun ; 97: 1-3, 2021 10.
Article in English | MEDLINE | ID: covidwho-1330652

ABSTRACT

The rapid development and application of different SARS-Cov2 vaccines world-wide has resulted in impressive efficacy and protection from this deadly pandemic. However, the existence of different and continuously developing vaccine candidates coupled with the likelihood of continued application due to both waning immune responses and emergence of viral mutants, means that more basic research regarding their efficacy and continued application are needed. This is particularly true with use of preclinical models involving effects when given during pregnancy. The substantial body of data on the impact of maternal immune activation (MIA) on neurologic development and behavior in the progeny necessitates the need to have all vaccine candidates, particularly when inducing strong toll receptor (TLR) responses, involving these models. Use of other preclinical models involving autoimmunity and allergy coupled with incorporation of human modifying variables of aging and obesity should also be applied to better reflect the heterogeneity of the general population and potential off-target effects that may arise. Additionally, the use of human ACE2 receptor transgenic mouse models can shed insights given the differential tissues expression at different stages in development. However, to foster these types of basic research studies involving different vaccine products, initiatives must first be implemented and supported at the governmental level even while clinical data still accumulates.


Subject(s)
COVID-19 , Vaccines , Animals , Female , Humans , Mice , Pandemics , Pregnancy , RNA, Viral , SARS-CoV-2
12.
FEBS J ; 289(12): 3374-3392, 2022 06.
Article in English | MEDLINE | ID: covidwho-1231088

ABSTRACT

Intragravidic and perinatal infections, acting through either direct viral effect or immune-mediated responses, are recognized causes of liability for neurodevelopmental disorders in the progeny. The large amounts of epidemiological data and the wealth of information deriving from animal models of gestational infections have contributed to delineate, in the last years, possible underpinning mechanisms for this phenomenon, including defects in neuronal migration, impaired spine and synaptic development, and altered activation of microglia. Recently, dysfunctions of the neurovascular unit and anomalies of the brain vasculature have unexpectedly emerged as potential causes at the origin of behavioral abnormalities and psychiatric disorders consequent to prenatal and perinatal infections. This review aims to discuss the up-to-date literature evidence pointing to the neurovascular unit and brain vasculature damages as the etiological mechanisms in neurodevelopmental syndromes. We focus on the inflammatory events consequent to intragravidic viral infections as well as on the direct viral effects as the potential primary triggers. These authors hope that a timely review of the literature will help to envision promising research directions, also relevant for the present and future COVID-19 longitudinal studies.


Subject(s)
COVID-19 , Mental Disorders , Pregnancy Complications, Infectious , Prenatal Exposure Delayed Effects , Animals , Brain , Female , Humans , Pregnancy
13.
Front Psychol ; 12: 614451, 2021.
Article in English | MEDLINE | ID: covidwho-1191717

ABSTRACT

The emergent Coronavirus Disease 2019 (COVID-19) caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) could produce a maternal immune activation (MIA) via the inflammatory response during gestation that may impair fetal neurodevelopment and lead to postnatal and adulthood mental illness and behavioral dysfunctions. However, so far, limited evidence exists regarding long-term physiological, immunological, and neurodevelopmental modifications produced by the SARS-CoV-2 in the human maternal-fetal binomial and, particularly, in the offspring. Relevant findings derived from epidemiological and preclinical models show that a MIA is indeed linked to an increased risk of neurodevelopmental disorders in the offspring. We hypothesize that a gestational infection triggered by SARS-CoV-2 increases the risks leading to neurodevelopmental disorders of the newborn, which can affect childhood and the long-term quality of life. In particular, disruption of either the maternal or the fetal cholinergic anti-inflammatory pathway (CAP) could cause or exacerbate the severity of COVID-19 in the maternal-fetal binomial. From a translational perspective, in this paper, we discuss the possible manifestation of a MIA by SARS-CoV-2 and the subsequent neurodevelopmental disorders considering the role of the fetal-maternal cytokine cross-talk and the CAP. Specifically, we highlight the urgent need of preclinical studies as well as multicenter and international databanks of maternal-fetal psychophysiological data obtained pre-, during, and post-infection by SARS-CoV-2 from pregnant women and their offspring.

14.
Early Hum Dev ; 155: 105322, 2021 04.
Article in English | MEDLINE | ID: covidwho-1053333

ABSTRACT

From the moment of the identification of SARS-CoV-2 as an etiological agent of the severe clinical pictures of pneumonia that were being slowly observed all over the world, numerous studies have been conducted to increase the knowledge about what was an unknown virus until then. The efforts were mainly aimed to acquire epidemiological, microbiological, pathogenetic, clinical, diagnostic, therapeutic and preventive information in order to increase the available weapons to fight an infection which was rapidly taking on the characteristics of the pandemic. Given the topicality of the problem, not everything has yet been fully understood and clarified, especially in the maternal-fetal­neonatal field, where we are beginning to question what could be the outcomes of newborn babies born to mothers who contracted SARS-CoV-2 infection during pregnancy. Thus, the aim of this review is to analyze the long-term outcomes of this infection that could affect the offspring, regardless of a possible maternal-fetal transmission, focusing on, above all, the role of maternal immune activation and the expression of the Angiotensin-converting enzyme 2 (ACE2) in particular at the placental level.


Subject(s)
COVID-19/complications , Infant, Newborn, Diseases/virology , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , Adaptive Immunity , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/immunology , COVID-19/therapy , COVID-19/transmission , Female , Fetal Development , Humans , Infant, Newborn , Infant, Newborn, Diseases/embryology , Infant, Newborn, Diseases/metabolism , Infectious Disease Transmission, Vertical , Placenta/immunology , Placenta/metabolism , Pregnancy , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL