Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 47
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.
Front Med (Lausanne) ; 10: 1129288, 2023.
Article in English | MEDLINE | ID: covidwho-2312721

ABSTRACT

Background: Symptoms lasting longer than 12 weeks after severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection are called post-coronavirus disease (COVID) syndrome (PCS). The identification of new biomarkers that predict the occurrence or course of PCS in terms of a post-viral syndrome is vital. T-cell dysfunction, cytokine imbalance, and impaired autoimmunity have been reported in PCS. Nevertheless, there is still a lack of conclusive information on the underlying mechanisms due to, among other things, a lack of controlled study designs. Methods: Here, we conducted a prospective, controlled study to characterize the humoral and cellular immune response in unvaccinated patients with and without PCS following SARS-CoV-2 infection over 7 months and unexposed donors. Results: Patients with PCS showed as early as 6 weeks and 7 months after symptom onset significantly increased frequencies of SARS-CoV-2-specific CD4+ and CD8+ T-cells secreting IFNγ, TNF, and expressing CD40L, as well as plasmacytoid dendritic cells (pDC) with an activated phenotype. Remarkably, the immunosuppressive counterparts type 1 regulatory T-cells (TR1: CD49b/LAG-3+) and IL-4 were more abundant in PCS+. Conclusion: This work describes immunological alterations between inflammation and immunosuppression in COVID-19 convalescents with and without PCS, which may provide potential directions for future epidemiological investigations and targeted treatments.

3.
Front Immunol ; 14: 1031336, 2023.
Article in English | MEDLINE | ID: covidwho-2300731

ABSTRACT

Hospitalized patients have an increased risk of developing hospital-acquired sacral pressure injury (HASPI). However, it is unknown whether SARS-CoV-2 infection affects HASPI development. To explore the role of SARS-CoV-2 infection in HASPI development, we conducted a single institution, multi-hospital, retrospective study of all patients hospitalized for ≥5 days from March 1, 2020 to December 31, 2020. Patient demographics, hospitalization information, ulcer characteristics, and 30-day-related morbidity were collected for all patients with HASPIs, and intact skin was collected from HASPI borders in a patient subset. We determined the incidence, disease course, and short-term morbidity of HASPIs in COVID-19(+) patients, and characterized the skin histopathology and tissue gene signatures associated with HASPIs in COVID-19 disease. COVID-19(+) patients had a 63% increased HASPI incidence rate, HASPIs of more severe ulcer stage (OR 2.0, p<0.001), and HASPIs more likely to require debridement (OR 3.1, p=0.04) compared to COVID-19(-) patients. Furthermore, COVID-19(+) patients with HASPIs had 2.2x increased odds of a more severe hospitalization course compared to COVID-19(+) patients without HASPIs. HASPI skin histology from COVID-19(+) patients predominantly showed thrombotic vasculopathy, with the number of thrombosed vessels being significantly greater than HASPIs from COVID-19(-) patients. Transcriptional signatures of a COVID-19(+) sample subset were enriched for innate immune responses, thrombosis, and neutrophil activation genes. Overall, our results suggest that immunologic dysregulation secondary to SARS-CoV-2 infection, including neutrophil dysfunction and abnormal thrombosis, may play a pathogenic role in development of HASPIs in patients with severe COVID-19.


Subject(s)
COVID-19 , Pressure Ulcer , Thrombosis , Humans , COVID-19/epidemiology , Pressure Ulcer/epidemiology , SARS-CoV-2 , Retrospective Studies , Ulcer , Neutrophil Activation , Incidence , Thrombosis/epidemiology , Thrombosis/etiology , Hospitals
5.
BMC Infect Dis ; 23(1): 97, 2023 Feb 16.
Article in English | MEDLINE | ID: covidwho-2265011

ABSTRACT

BACKGROUND: Individuals with post-acute sequelae of COVID (PASC) may have a persistence in immune activation that differentiates them from individuals who have recovered from COVID without clinical sequelae. To investigate how humoral immune activation may vary in this regard, we compared patterns of vaccine-provoked serological response in patients with PASC compared to individuals recovered from prior COVID without PASC. METHODS: We prospectively studied 245 adults clinically diagnosed with PASC and 86 adults successfully recovered from prior COVID. All participants had measures of humoral immunity to SARS-CoV-2 assayed before or after receiving their first-ever administration of COVID vaccination (either single-dose or two-dose regimen), including anti-spike (IgG-S and IgM-S) and anti-nucleocapsid (IgG-N) antibodies as well as IgG-S angiotensin-converting enzyme 2 (ACE2) binding levels. We used unadjusted and multivariable-adjusted regression analyses to examine the association of PASC compared to COVID-recovered status with post-vaccination measures of humoral immunity. RESULTS: Individuals with PASC mounted consistently higher post-vaccination IgG-S antibody levels when compared to COVID-recovered (median log IgG-S 3.98 versus 3.74, P < 0.001), with similar results seen for ACE2 binding levels (median 99.1 versus 98.2, P = 0.044). The post-vaccination IgM-S response in PASC was attenuated but persistently unchanged over time (P = 0.33), compared to in COVID recovery wherein the IgM-S response expectedly decreased over time (P = 0.002). Findings remained consistent when accounting for demographic and clinical variables including indices of index infection severity and comorbidity burden. CONCLUSION: We found evidence of aberrant immune response distinguishing PASC from recovered COVID. This aberrancy is marked by excess IgG-S activation and ACE2 binding along with findings consistent with a delayed or dysfunctional immunoglobulin class switching, all of which is unmasked by vaccine provocation. These results suggest that measures of aberrant immune response may offer promise as tools for diagnosing and distinguishing PASC from non-PASC phenotypes, in addition to serving as potential targets for intervention.


Subject(s)
COVID-19 Vaccines , COVID-19 , Post-Acute COVID-19 Syndrome , Humans , Angiotensin-Converting Enzyme 2 , Antibodies, Viral , COVID-19/prevention & control , Disease Progression , Immunoglobulin G , Immunoglobulin M , SARS-CoV-2 , Vaccination , Post-Acute COVID-19 Syndrome/immunology , COVID-19 Vaccines/immunology
6.
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
7.
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
8.
Vaccines (Basel) ; 11(2)2023 Feb 13.
Article in English | MEDLINE | ID: covidwho-2242727

ABSTRACT

BACKGROUND: It is widely accepted that SARS-CoV-2 causes a dysregulation of immune and coagulation processes. In severely affected patients, viral sepsis may result in life endangering multiple organ dysfunction. Furthermore, most therapies for COVID-19 patients target either the immune system or coagulation processes. As the exact mechanism causing SARS-CoV-2-induced morbidity and mortality was unknown, we started an in-depth analysis of immunologic and coagulation processes. METHODS: 127 COVID-19 patients were treated at the University Hospital Essen, Germany, between May 2020 and February 2022. Patients were divided according to their maximum COVID-19 WHO ordinal severity score (WHO 0-10) into hospitalized patients with a non-severe course of disease (WHO 4-5, n = 52) and those with a severe course of disease (WHO 6-10, n = 75). Non-infected individuals served as healthy controls (WHO 0, n = 42). Blood was analyzed with respect to cell numbers, clotting factors, as well as pro- and anti-inflammatory mediators in plasma. As functional parameters, phagocytosis and inflammatory responses to LPS and antigen-specific stimulation were determined in monocytes, granulocytes, and T cells using flow cytometry. FINDINGS: In the present study, immune and coagulation systems were analyzed simultaneously. Interestingly, many severe COVID-19 patients showed an upregulation of pro-inflammatory mediators and at the same time clear signs of immunosuppression. Furthermore, severe COVID-19 patients not only exhibited a disturbed immune system, but in addition showed a pronounced pro-coagulation phenotype with impaired fibrinolysis. Therefore, our study adds another puzzle piece to the already complex picture of COVID-19 pathology implying that therapies in COVID-19 must be individualized. CONCLUSION: Despite years of research, COVID-19 has not been understood completely and still no therapies exist, fitting all requirements and phases of COVID-19 disease. This observation is highly reminiscent to sepsis. Research in sepsis has been going on for decades, while the disease is still not completely understood and therapies fitting all patients are lacking as well. In both septic and COVID-19 patients, immune activation can be accompanied by immune paralysis, complicating therapeutic intervention. Accordingly, therapies that lower immune activation may cause detrimental effects in patients, who are immune paralyzed by viral infections or sepsis. We therefore suggest individualizing therapies and to broaden the spectrum of immunological parameters analyzed before therapy. Only if the immune status of a patient is understood, can a therapeutic intervention be successful.

9.
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.

10.
RNA Technologies ; 13:237-255, 2022.
Article in English | Scopus | ID: covidwho-2059698

ABSTRACT

Messenger RNA (mRNA) can be harnessed as vaccines and therapeutic drugs via transient in situ expression of protein antigens and therapeutic proteins, respectively. Currently, mRNA-based vaccines are used worldwide in mass vaccination programs to induce protective immunity against COVID-19, and a number of prophylactic vaccines, therapeutic vaccines, and therapeutic drugs based on mRNA are now tested in clinical trials. Although chemical modification of the mRNA components has considerably ameliorated mRNA stability and immunogenicity, further improvements in formulation and delivery systems, which are used to transport mRNA to the cytosol of target cells, are still required to enhance the efficacy and safety of mRNA therapeutics. However, our knowledge about the mechanisms by which mRNA therapeutics activate the immune system is still very limited, partly because the activation of immune cells by ionizable lipids commonly used in mRNA delivery systems is poorly understood. Lipid-mediated induction of innate immune pathways can be exploited in mRNA vaccines by providing an adjuvant effect, whereas innate immune activation is undesired for the therapeutic use of mRNA. Here, we review recent studies focusing on the hurdles that challenge in vivo delivery of mRNA. We subsequently discuss the state of the art in formulation design approaches, which are used to overcome these challenges, with focus on the marketed COVID-19 mRNA vaccines. Finally, we present research centered on how ionizable and cationic lipids used for delivery of mRNA therapeutics activate immune cells and engage innate immune pathways, including future challenges and opportunities in formulation and delivery to optimize the safe and efficacious use of mRNA therapeutics. © 2022, The Author(s), under exclusive license to Springer Nature Switzerland AG.

11.
Front Immunol ; 13: 815041, 2022.
Article in English | MEDLINE | ID: covidwho-1952315

ABSTRACT

The role of T cells in the control of SARS-CoV-2 infection has been underestimated in favor of neutralizing antibodies. However, cellular immunity is essential for long-term viral control and protection from disease severity. To understand T-cell immunity in the absence of antibody generation we focused on a group of SARS-CoV-2 Non-Seroconvertors (NSC) recovered from infection. We performed an immune comparative analysis of SARS-CoV-2 infected individuals stratified by the absence or presence of seroconversion and disease severity. We report high levels of total naïve and low effector CD8+ T cells in NSC. Moreover, reduced levels of T-cell activation monitored by PD-1 and activation-induced markers were observed in the context of functional SARS-CoV-2 T-cell responses. Longitudinal data indicate the stability of the NSC phenotype over three months of follow-up after infection. Together, these data characterized distinctive immunological traits in NSC including skewed cellular distribution, low activation and functional SARS-CoV-2 T-cell responses. This data highlights the value of T-cell immune monitoring in populations with low seroconversion rates in response to SARS-CoV-2 infection and vaccination.


Subject(s)
COVID-19 , T-Lymphocytes , Humans , Immunity, Cellular , SARS-CoV-2 , Vaccination
12.
Front Neurosci ; 16: 943903, 2022.
Article in English | MEDLINE | ID: covidwho-1917227

ABSTRACT

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

13.
Front Immunol ; 12: 785941, 2021.
Article in English | MEDLINE | ID: covidwho-1869377

ABSTRACT

Coronavirus disease 2019 (COVID-19) is an infectious disease caused by beta-coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that has rapidly spread across the globe starting from February 2020. It is well established that during viral infection, extracellular vesicles become delivery/presenting vectors of viral material. However, studies regarding extracellular vesicle function in COVID-19 pathology are still scanty. Here, we performed a comparative study on exosomes recovered from the plasma of either MILD or SEVERE COVID-19 patients. We show that although both types of vesicles efficiently display SARS-CoV-2 spike-derived peptides and carry immunomodulatory molecules, only those of MILD patients are capable of efficiently regulating antigen-specific CD4+ T-cell responses. Accordingly, by mass spectrometry, we show that the proteome of exosomes of MILD patients correlates with a proper functioning of the immune system, while that of SEVERE patients is associated with increased and chronic inflammation. Overall, we show that exosomes recovered from the plasma of COVID-19 patients possess SARS-CoV-2-derived protein material, have an active role in enhancing the immune response, and possess a cargo that reflects the pathological state of patients in the acute phase of the disease.


Subject(s)
Adaptive Immunity , COVID-19/immunology , Exosomes/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Acute Disease , Adult , Aged , COVID-19/blood , Exosomes/metabolism , Female , Humans , Male , Middle Aged , Plasma , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/blood
14.
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
15.
Respir Res ; 23(1): 127, 2022 May 18.
Article in English | MEDLINE | ID: covidwho-1849735

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is characterized by persistent airflow limitation associated with chronic inflammation in the airways. Mucosal-associated invariant T (MAIT) cells are unconventional, innate-like T cells highly abundant in mucosal tissues including the lung. We hypothesized that the characteristics of MAIT cells in circulation may be prospectively associated with COPD morbidity. METHODS: COPD subjects (n = 61) from the Tools for Identifying Exacerbations (TIE) study were recruited when in stable condition. At study entry, forced expiratory volume in 1 s (FEV1) was measured and peripheral blood mononuclear cells were cryopreserved for later analysis by flow cytometry. Patients were followed for 3 years to record clinically meaningful outcomes. RESULTS: Patients who required hospitalization at one or more occasions during the 3-year follow-up (n = 21) had lower MAIT cell counts in peripheral blood at study inclusion, compared with patients who did not get hospitalized (p = 0.036). In contrast, hospitalized and never hospitalized patients did not differ in CD8 or CD4 T cell counts (p = 0.482 and p = 0.221, respectively). Moreover, MAIT cells in hospitalized subjects showed a more activated phenotype with higher CD38 expression (p = 0.014), and there was a trend towards higher LAG-3 expression (p = 0.052). Conventional CD4 and CD8 T cells were similar between the groups. Next we performed multi-variable logistic regression analysis with hospitalizations as dependent variable, and FEV1, GOLD 2017 group, and quantity or activation of MAIT and conventional T cells as independent variables. MAIT cell count, CD38 expression on MAIT cells, and LAG-3 expression on both MAIT and CD8 T cells were all independently associated with the risk of hospitalization. CONCLUSIONS: These findings suggest that MAIT cells might reflect a novel, FEV1-independent immunological dimension in the complexity of COPD. The potential implication of MAIT cells in COPD pathogenesis and MAIT cells' prognostic potential deserve further investigation.


Subject(s)
Mucosal-Associated Invariant T Cells , Pulmonary Disease, Chronic Obstructive , Hospitalization , Humans , Leukocytes, Mononuclear , Lymphocyte Count , Pulmonary Disease, Chronic Obstructive/metabolism
16.
Vaccines (Basel) ; 10(5)2022 Apr 22.
Article in English | MEDLINE | ID: covidwho-1810358

ABSTRACT

Mucormycosis is a group of infections, caused by multiple fungal species, which affect many human organs and is lethal in immunocompromised patients. During the COVID-19 pandemic, the current wave of mucormycosis is a challenge to medical professionals as its effects are multiplied because of the severity of COVID-19 infection. The variant of concern, Omicron, has been linked to fatal mucormycosis infections in the US and Asia. Consequently, current postdiagnostic treatments of mucormycosis have been rendered unsatisfactory. In this hour of need, a preinfection cure is needed that may prevent lethal infections in immunocompromised individuals. This study proposes a potential vaccine construct targeting mucor and rhizopus species responsible for mucormycosis infections, providing immunoprotection to immunocompromised patients. The vaccine construct, with an antigenicity score of 0.75 covering, on average, 92-98% of the world population, was designed using an immunoinformatics approach. Molecular interactions with major histocompatibility complex-1 (MHC-I), Toll-like receptors-2 (TLR2), and glucose-regulated protein 78 (GRP78), with scores of -896.0, -948.4, and -925.0, respectively, demonstrated its potential to bind with the human immune receptors. It elicited a strong predicted innate and adaptive immune response in the form of helper T (Th) cells, cytotoxic T (TC) cells, B cells, natural killer (NK) cells, and macrophages. The vaccine cloned in the pBR322 vector showed positive amplification, further solidifying its stability and potential. The proposed construct holds a promising approach as the first step towards an antimucormycosis vaccine and may contribute to minimizing postdiagnostic burdens and failures.

17.
Trends Genet ; 38(7): 662-675, 2022 07.
Article in English | MEDLINE | ID: covidwho-1778469

ABSTRACT

Maternal immune activation (MIA) and infection during pregnancy are known to reprogramme offspring phenotypes. However, the epigenetic effects of preconceptual paternal infection and paternal immune activation (PIA) are not currently well understood. Recent reports show that paternal infection and immune activation can affect offspring phenotypes, particularly brain function, behaviour, and immune system functioning, across multiple generations without re-exposure to infection. Evidence from other environmental exposures indicates that epigenetic inheritance also occurs in humans. Given the growing impact of the coronavirus disease 2019 (COVID-19) pandemic, it is imperative that we investigate all of the potential epigenetic mechanisms and multigenerational phenotypes that may arise from both maternal and paternal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, as well as associated MIA, PIA, and inflammation. This will allow us to understand and, if necessary, mitigate any potential changes in disease susceptibility in the children, and grandchildren, of affected parents.


Subject(s)
COVID-19 , SARS-CoV-2 , COVID-19/genetics , Disease Susceptibility , Epigenesis, Genetic , Female , Humans , Parents , Pregnancy , SARS-CoV-2/genetics
18.
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
19.
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.

20.
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
SELECTION OF CITATIONS
SEARCH DETAIL