Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Viruses ; 15(5)2023 05 14.
Article in English | MEDLINE | ID: covidwho-20232730

ABSTRACT

Chikungunya virus (CHIKV) and Zika virus (ZIKV) are important disease-causing agents worldwide. Currently, there are no antiviral drugs or vaccines approved to treat these viruses. However, peptides have shown great potential for new drug development. A recent study described (p-BthTX-I)2K [(KKYRYHLKPF)2K], a peptide derived from the Bothropstoxin-I toxin in the venom of the Bothrops jararacussu snake, showed antiviral activity against SARS-CoV-2. In this study, we assessed the activity of this peptide against CHIKV and ZIKV and its antiviral action in the different stages of the viral replication cycle in vitro. We observed that (p-BthTX-I)2K impaired CHIKV infection by interfering with the early steps of the viral replication cycle, reducing CHIKV entry into BHK-21 cells specifically by reducing both the attachment and internalization steps. (p-BthTX-I)2K also inhibited the ZIKV replicative cycle in Vero cells. The peptide protected the cells against ZIKV infection and decreased the levels of the viral RNA and the NS3 protein of this virus at viral post-entry steps. In conclusion, this study highlights the potential of the (p-BthTX-I)2K peptide to be a novel broad-spectrum antiviral candidate that targets different steps of the replication cycle of both CHIKV and ZIKV.


Subject(s)
COVID-19 , Chikungunya Fever , Chikungunya virus , Viruses , Zika Virus Infection , Zika Virus , Animals , Chlorocebus aethiops , Humans , Zika Virus Infection/drug therapy , Zika Virus/genetics , Vero Cells , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Virus Replication , SARS-CoV-2 , Chikungunya virus/genetics , Peptides/pharmacology , Peptides/therapeutic use
2.
Methods Mol Biol ; 2621: 307-323, 2023.
Article in English | MEDLINE | ID: covidwho-2297362

ABSTRACT

Zika virus (ZIKV) infection may cause serious birth defects and is a critical concern for women of child-bearing age in affected regions. A simple, portable, and easy-to-use ZIKV detection method would enable point-of-care testing, which may aid in prevention of the spread of the virus. Herein, we describe a reverse transcription isothermal loop-mediated amplification (RT-LAMP) method that detects the presence of ZIKV RNA in complex samples (e.g., blood, urine, and tap water). Phenol red is the colorimetric indicator of successful amplification. Color changes based on the amplified RT-LAMP product from the presence of viral target are monitored using a smartphone camera under ambient light conditions. A single viral RNA molecule per µL can be detected in as quickly as 15 min using this method with 100% sensitivity and 100% specificity in blood and tap water, while 100% sensitivity and 67% specificity in urine. This platform can also be used to identify other viruses including SARS-CoV-2 and improve the current state of field-based diagnostics.


Subject(s)
COVID-19 , Zika Virus Infection , Zika Virus , Female , Humans , Zika Virus/genetics , Microfluidics , Smartphone , Sensitivity and Specificity , SARS-CoV-2
3.
J Neurovirol ; 29(2): 121-134, 2023 04.
Article in English | MEDLINE | ID: covidwho-2304443

ABSTRACT

Progress in stem cell research has revolutionized the medical field for more than two decades. More recently, the discovery of induced pluripotent stem cells (iPSCs) has allowed for the development of advanced disease modeling and tissue engineering platforms. iPSCs are generated from adult somatic cells by reprogramming them into an embryonic-like state via the expression of transcription factors required for establishing pluripotency. In the context of the central nervous system (CNS), iPSCs have the potential to differentiate into a wide variety of brain cell types including neurons, astrocytes, microglial cells, endothelial cells, and oligodendrocytes. iPSCs can be used to generate brain organoids by using a constructive approach in three-dimensional (3D) culture in vitro. Recent advances in 3D brain organoid modeling have provided access to a better understanding of cell-to-cell interactions in disease progression, particularly with neurotropic viral infections. Neurotropic viral infections have been difficult to study in two-dimensional culture systems in vitro due to the lack of a multicellular composition of CNS cell networks. In recent years, 3D brain organoids have been preferred for modeling neurotropic viral diseases and have provided invaluable information for better understanding the molecular regulation of viral infection and cellular responses. Here we provide a comprehensive review of the literature on recent advances in iPSC-derived 3D brain organoid culturing and their utilization in modeling major neurotropic viral infections including HIV-1, HSV-1, JCV, ZIKV, CMV, and SARS-CoV2.


Subject(s)
COVID-19 , Induced Pluripotent Stem Cells , Virus Diseases , Viruses , Zika Virus Infection , Zika Virus , Humans , Induced Pluripotent Stem Cells/metabolism , Zika Virus Infection/genetics , Endothelial Cells , RNA, Viral/metabolism , SARS-CoV-2 , Brain , Virus Diseases/metabolism , Organoids/metabolism
4.
Wellcome Open Res ; 6: 241, 2021.
Article in English | MEDLINE | ID: covidwho-2293550

ABSTRACT

Emerging and re-emerging viruses are a global health concern. Genome sequencing as an approach for monitoring circulating viruses is currently hampered by complex and expensive methods. Untargeted, metagenomic nanopore sequencing can provide genomic information to identify pathogens, prepare for or even prevent outbreaks. SMART (Switching Mechanism at the 5' end of RNA Template) is a popular approach for RNA-Seq but most current methods rely on oligo-dT priming to target polyadenylated mRNA molecules. We have developed two random primed SMART-Seq approaches, a sequencing agnostic approach 'SMART-9N' and a version compatible rapid adapters  available from Oxford Nanopore Technologies 'Rapid SMART-9N'. The methods were developed using viral isolates, clinical samples, and compared to a gold-standard amplicon-based method. From a Zika virus isolate the SMART-9N approach recovered 10kb of the 10.8kb RNA genome in a single nanopore read. We also obtained full genome coverage at a high depth coverage using the Rapid SMART-9N, which takes only 10 minutes and costs up to 45% less than other methods. We found the limits of detection of these methods to be 6 focus forming units (FFU)/mL with 99.02% and 87.58% genome coverage for SMART-9N and Rapid SMART-9N respectively. Yellow fever virus plasma samples and SARS-CoV-2 nasopharyngeal samples previously confirmed by RT-qPCR with a broad range of Ct-values were selected for validation. Both methods produced greater genome coverage when compared to the multiplex PCR approach and we obtained the longest single read of this study (18.5 kb) with a SARS-CoV-2 clinical sample, 60% of the virus genome using the Rapid SMART-9N method. This work demonstrates that SMART-9N and Rapid SMART-9N are sensitive, low input, and long-read compatible alternatives for RNA virus detection and genome sequencing and Rapid SMART-9N improves the cost, time, and complexity of laboratory work.

5.
Exp Neurol ; 363: 114375, 2023 05.
Article in English | MEDLINE | ID: covidwho-2271639

ABSTRACT

Microglia, the resident macrophage of the central nervous system, are increasingly recognized as contributing to diverse aspects of human development, health, and disease. In recent years, numerous studies in both mouse and human models have identified microglia as a "double edged sword" in the progression of neurotropic viral infections: protecting against viral replication and cell death in some contexts, while acting as viral reservoirs and promoting excess cellular stress and cytotoxicity in others. It is imperative to understand the diversity of human microglial responses in order to therapeutically modulate them; however, modeling human microglia has been historically challenging due to significant interspecies differences in innate immunity and rapid transformation upon in vitro culture. In this review, we discuss the contribution of microglia to the neuropathogenesis of key neurotropic viral infections: human immunodeficiency virus 1 (HIV-1), Zika virus (ZIKV), Japanese encephalitis virus (JEV), West Nile virus (WNV), Herpes simplex virus (HSV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We pay special attention to recent work with human stem cell-derived microglia and propose strategies to leverage these powerful models to further uncover species- and disease-specific microglial responses and novel therapeutic interventions for neurotropic viral infections.


Subject(s)
COVID-19 , Zika Virus Infection , Zika Virus , Humans , Animals , Mice , Microglia/metabolism , Host Microbial Interactions , Zika Virus Infection/metabolism , COVID-19/metabolism , SARS-CoV-2
6.
Neuromethods ; 189:173-190, 2023.
Article in English | EMBASE | ID: covidwho-2059680

ABSTRACT

In recent years, we are living through different viral pandemics that result in neurological impairments. Given the human-specific nature of brain development, physiology, and pathology, it is imperative to use human models to investigate the neurological impact of viral infections, such as Zika virus and SARS-CoV-2. Brain organoids are powerful in vitro platforms for the analysis of the effects of viral infections on brain development and function, with prospective application to new emerging viral threats. Using brain organoids, it was possible to show that Zika virus infects neural stem cells, disrupting the cell cycle and neurogenesis, leading to microcephaly, a severe reduction of the brain. On the other hand, while it is still under investigation how SARS-CoV-2 might enter and alter the brain, organoid studies are helping to characterize its neurotropism and potential mechanisms of neurovirulence. Here, we describe a method for the infection of human brain organoid cultures with Zika and Sars-CoV-2 viruses that can be used to study neurodevelopmental phenotypes, alteration in neuronal functionality, host-pathogen interactions, as well as for drug testing. Copyright © 2023, Springer Science+Business Media, LLC, part of Springer Nature.

7.
Front Immunol ; 13: 905431, 2022.
Article in English | MEDLINE | ID: covidwho-1883914

ABSTRACT

The Zika virus (ZIKV) epidemic poses a substantial threat to the public, and the development of safe and effective vaccines is a demanding challenge. In this study, we constructed a kind of self-assembling nanovaccine which confers complete protection against ZIKV infection. The ZIKV envelop protein domain III (zEDIII) was presented on recombinant human heavy chain ferritin (rHF) to form the zEDIII-rHF nanoparticle. Immunization of mice with zEDIII-rHF nanoparticle in the absence of an adjuvant induced robust humoral and cellular immune responses. zEDIII-rHF vaccination conferred complete protection against lethal infection with ZIKV and eliminated pathological symptoms in the brain. Importantly, the zEDIII-rHF nanovaccine induced immune response did not cross-react with dengue virus-2, overcoming the antibody-dependent enhancement (ADE) problem that is a safety concern for ZIKV vaccine development. Our constructed zEDIII-rHF nanovaccine, with superior protective performance and avoidance of ADE, provides an effective and safe vaccine candidate against ZIKV.


Subject(s)
Zika Virus Infection , Zika Virus , Animals , Antibodies, Viral , Antibody-Dependent Enhancement , Immunization , Mice
8.
Viruses ; 14(3)2022 03 18.
Article in English | MEDLINE | ID: covidwho-1760845

ABSTRACT

Pathogenesis of viral infections of the central nervous system (CNS) is poorly understood, and this is partly due to the limitations of currently used preclinical models. Brain organoid models can overcome some of these limitations, as they are generated from human derived stem cells, differentiated in three dimensions (3D), and can mimic human neurodevelopmental characteristics. Therefore, brain organoids have been increasingly used as brain models in research on various viruses, such as Zika virus, severe acute respiratory syndrome coronavirus 2, human cytomegalovirus, and herpes simplex virus. Brain organoids allow for the study of viral tropism, the effect of infection on organoid function, size, and cytoarchitecture, as well as innate immune response; therefore, they provide valuable insight into the pathogenesis of neurotropic viral infections and testing of antivirals in a physiological model. In this review, we summarize the results of studies on viral CNS infection in brain organoids, and we demonstrate the broad application and benefits of using a human 3D model in virology research. At the same time, we describe the limitations of the studies in brain organoids, such as the heterogeneity in organoid generation protocols and age at infection, which result in differences in results between studies, as well as the lack of microglia and a blood brain barrier.


Subject(s)
COVID-19 , Central Nervous System Viral Diseases , Zika Virus Infection , Zika Virus , Blood-Brain Barrier , Brain/pathology , Humans , Organoids , Zika Virus Infection/pathology
9.
Front Immunol ; 13: 826091, 2022.
Article in English | MEDLINE | ID: covidwho-1731778

ABSTRACT

Neural stem cells (NSCs) are multipotent stem cells that reside in the fetal and adult mammalian brain, which can self-renew and differentiate into neurons and supporting cells. Intrinsic and extrinsic cues, from cells in the local niche and from distant sites, stringently orchestrates the self-renewal and differentiation competence of NSCs. Ample evidence supports the important role of NSCs in neuroplasticity, aging, disease, and repair of the nervous system. Indeed, activation of NSCs or their transplantation into injured areas of the central nervous system can lead to regeneration in animal models. Viral invasion of NSCs can negatively affect neurogenesis and synaptogenesis, with consequent cell death, impairment of cell cycle progression, early differentiation, which cause neural progenitors depletion in the cortical layer of the brain. Herein, we will review the current understanding of Zika virus (ZIKV) infection of the fetal brain and the NSCs, which are the preferential population targeted by ZIKV. Furthermore, the potential neurotropic properties of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which may cause direct neurological damage, will be discussed.


Subject(s)
Brain/virology , COVID-19/pathology , COVID-19/virology , Neurogenesis/physiology , Neurons/virology , Zika Virus Infection/pathology , Zika Virus Infection/virology , Animals , Humans , Neural Stem Cells/virology
10.
Front Public Health ; 9: 802428, 2021.
Article in English | MEDLINE | ID: covidwho-1636471

ABSTRACT

Introduction: Evidence on authorship trends of health research conducted about or in Africa shows that there is a lack of local researchers in the first and last authorship positions, with high income country collaborations taking up these positions. The differences in authorship calls into question power imbalances in global health research and who benefits from the production of new discoveries and innovations. Health studies may further go on to inform policy and clinical practice within the region having an impact on public health. This paper aims to compare the differences in authorship between COVID-19 and relevant infectious diseases in Africa. Materials and Methods: We will conduct a bibliometric analysis comparing authorship for COVID-19 research during a public health emergency with authorship for four other infectious diseases of relevance to Africa namely: Ebola, Zika Virus (ZIKV), Tuberculosis (TB) and Influenza. Our scoping review will follow the framework developed by Arksey and O'Malley and reviewed by Levac et al. We will search MEDLINE (Ovid), African Index Medicus (AIM), Eastern Mediterranean Region (IMEMR) Index Medicus, Embase (Ovid), and Web of Science (Clarivate). We will compare the different trends of disease research between the selected diseases. This study is registered with OSF registries and is licensed with the Academic Free License version 3.0. The open science registration number is 10.17605/OSF.IO/5ZPGN.


Subject(s)
COVID-19 , Zika Virus Infection , Zika Virus , Africa , Bibliometrics , Delivery of Health Care , Humans , Review Literature as Topic , SARS-CoV-2
11.
Vaccines (Basel) ; 9(12)2021 Dec 10.
Article in English | MEDLINE | ID: covidwho-1572684

ABSTRACT

Zika virus (ZIKV), a mosquito-borne flavivirus, has recently triggered global concern due to severe health complications. In 2015, a large ZIKV outbreak occurred in the Americas and established a link between ZIKV and microcephaly in newborn babies, spontaneous abortion, persistent viremia, and Guillain-Barré syndrome. While antivirals are being developed and prevention strategies focus on vector control, a safe and effective Zika vaccine remains unavailable. Messenger RNA (mRNA) vaccine technology has arisen as a flexible, simplified, and fast vaccine production platform. Here, we report on an mRNA vaccine candidate that encodes the pre-membrane and envelope (prM-E) glycoproteins of ZIKV strain Brazil SPH2015 and is encapsulated in lipid nanoparticles (LNPs). Our ZIKV prM-E mRNA-LNP vaccine candidate induced antibody responses that protected in AG129 mice deficient in interferon (IFN) alpha/beta/gamma (IFN-α/ß/γ) receptors. Notably, a single administration of ZIKV prM-E mRNA-LNP protected against a lethal dose of ZIKV, while a two-dose strategy induced strong protective immunity. E-specific double-positive IFN-γ and TNF-α T-cells were induced in BALB/c mice after immunizations with a two-dose strategy. With the success of mRNA vaccine technology in facing the coronavirus (COVID-19) pandemic, our data support the development of prM-E RNActive® as a promising mRNA vaccine against Zika to counter future epidemics.

12.
Viruses ; 13(11)2021 10 20.
Article in English | MEDLINE | ID: covidwho-1538529

ABSTRACT

Viruses are obligate parasites that depend on a host cell for replication and survival. Consequently, to fully understand the viral processes involved in infection and replication, it is fundamental to study them in the cellular context. Often, viral infections induce significant changes in the subcellular organization of the host cell due to the formation of viral factories, alteration of cell cytoskeleton and/or budding of newly formed particles. Accurate 3D mapping of organelle reorganization in infected cells can thus provide valuable information for both basic virus research and antiviral drug development. Among the available techniques for 3D cell imaging, cryo-soft X-ray tomography stands out for its large depth of view (allowing for 10 µm thick biological samples to be imaged without further thinning), its resolution (about 50 nm for tomographies, sufficient to detect viral particles), the minimal requirements for sample manipulation (can be used on frozen, unfixed and unstained whole cells) and the potential to be combined with other techniques (i.e., correlative fluorescence microscopy). In this review we describe the fundamentals of cryo-soft X-ray tomography, its sample requirements, its advantages and its limitations. To highlight the potential of this technique, examples of virus research performed at BL09-MISTRAL beamline in ALBA synchrotron are also presented.


Subject(s)
Tomography, X-Ray/methods , Virus Diseases/virology , Virus Physiological Phenomena , Animals , Antiviral Agents/pharmacology , Humans , Tomography, X-Ray/instrumentation , Virus Diseases/diagnostic imaging , Virus Diseases/drug therapy , Viruses/chemistry , Viruses/drug effects
13.
Viruses ; 13(11)2021 11 11.
Article in English | MEDLINE | ID: covidwho-1534242

ABSTRACT

CRISPR/Cas is a powerful tool for studying the role of genes in viral infections. The invention of CRISPR screening technologies has made it possible to untangle complex interactions between the host and viral agents. Moreover, whole-genome and pathway-specific CRISPR screens have facilitated identification of novel drug candidates for treating viral infections. In this review, we highlight recent developments in the fields of CRISPR/Cas with a focus on the use of CRISPR screens for studying viral infections and identifying new candidate genes to aid development of antivirals.


Subject(s)
CRISPR-Cas Systems , Genetic Techniques , Genome-Wide Association Study/methods , High-Throughput Screening Assays/methods , Virus Diseases/genetics , Virus Diseases/virology , Viruses/genetics , Drug Discovery , Host Microbial Interactions , Humans
14.
J Virol ; 95(24): e0059621, 2021 11 23.
Article in English | MEDLINE | ID: covidwho-1443352

ABSTRACT

Cellular factors have important roles in all facets of the flavivirus replication cycle. Deciphering viral-host protein interactions is essential for understanding the flavivirus life cycle as well as development of effective antiviral strategies. To uncover novel host factors that are co-opted by multiple flaviviruses, a CRISPR/Cas9 genome wide knockout (KO) screen was employed to identify genes required for replication of Zika virus (ZIKV). Receptor for Activated Protein C Kinase 1 (RACK1) was identified as a novel host factor required for ZIKV replication, which was confirmed via complementary experiments. Depletion of RACK1 via siRNA demonstrated that RACK1 is important for replication of a wide range of mosquito- and tick-borne flaviviruses, including West Nile Virus (WNV), Dengue Virus (DENV), Powassan Virus (POWV) and Langat Virus (LGTV) as well as the coronavirus SARS-CoV-2, but not for YFV, EBOV, VSV or HSV. Notably, flavivirus replication was only abrogated when RACK1 expression was dampened prior to infection. Utilising a non-replicative flavivirus model, we show altered morphology of viral replication factories and reduced formation of vesicle packets (VPs) in cells lacking RACK1 expression. In addition, RACK1 interacted with NS1 protein from multiple flaviviruses; a key protein for replication complex formation. Overall, these findings reveal RACK1's crucial role to the biogenesis of pan-flavivirus replication organelles. IMPORTANCE Cellular factors are critical in all facets of viral lifecycles, where overlapping interactions between the virus and host can be exploited as possible avenues for the development of antiviral therapeutics. Using a genome-wide CRISPR knockout screening approach to identify novel cellular factors important for flavivirus replication we identified RACK1 as a pro-viral host factor for both mosquito- and tick-borne flaviviruses in addition to SARS-CoV-2. Using an innovative flavivirus protein expression system, we demonstrate for the first time the impact of the loss of RACK1 on the formation of viral replication factories known as 'vesicle packets' (VPs). In addition, we show that RACK1 can interact with numerous flavivirus NS1 proteins as a potential mechanism by which VP formation can be induced by the former.


Subject(s)
CRISPR-Cas Systems , Flavivirus/genetics , Neoplasm Proteins/genetics , Receptors for Activated C Kinase/genetics , Virus Replication , A549 Cells , Aedes , Animals , COVID-19 , Chlorocebus aethiops , Culicidae , Dengue Virus/genetics , Genome-Wide Association Study , HEK293 Cells , Host-Pathogen Interactions/genetics , Humans , RNA, Small Interfering/metabolism , RNA, Viral/metabolism , SARS-CoV-2 , Vero Cells , West Nile virus/genetics , Zika Virus/genetics , Zika Virus Infection/virology
15.
Int J Environ Res Public Health ; 18(1)2021 01 02.
Article in English | MEDLINE | ID: covidwho-1389357

ABSTRACT

Infectious diseases have caused some of the most feared plagues and greatly harmed human health. However, despite the qualitative understanding that the occurrence and diffusion of infectious disease is related to the environment, the quantitative relations are unknown for many diseases. Zika virus (ZIKV) is a mosquito-borne virus that poses a fatal threat and has spread explosively throughout the world, impacting human health. From a geographical perspective, this study aims to understand the global hotspots of ZIKV as well as the spatially heterogeneous relationship between ZIKV and environmental factors using exploratory special data analysis (ESDA) model. A geographically weighted regression (GWR) model was used to analyze the influence of the dominant environmental factors on the spread of ZIKV at the continental scale. The results indicated that ZIKV transmission had obvious regional and seasonal heterogeneity. Population density, GDP per capita, and landscape fragmentation were the dominant environmental factors affecting the spread of ZIKV, which indicates that social factors had a greater influence than natural factors on the spread of it. As SARS-CoV-2 is spreading globally, this study can provide methodological reference for fighting against the pandemic.


Subject(s)
Zika Virus Infection , Animals , Humans , Mosquito Vectors , Spatio-Temporal Analysis , Zika Virus , Zika Virus Infection/epidemiology , Zika Virus Infection/transmission
16.
EMBO Rep ; 22(11): e52948, 2021 11 04.
Article in English | MEDLINE | ID: covidwho-1381494

ABSTRACT

The type I interferon (IFN) response is the major host arsenal against invading viruses. IRGM is a negative regulator of IFN responses under basal conditions. However, the role of human IRGM during viral infection has remained unclear. In this study, we show that IRGM expression is increased upon viral infection. IFN responses induced by viral PAMPs are negatively regulated by IRGM. Conversely, IRGM depletion results in a robust induction of key viral restriction factors including IFITMs, APOBECs, SAMHD1, tetherin, viperin, and HERC5/6. Additionally, antiviral processes such as MHC-I antigen presentation and stress granule signaling are enhanced in IRGM-deficient cells, indicating a robust cell-intrinsic antiviral immune state. Consistently, IRGM-depleted cells are resistant to the infection with seven viruses from five different families, including Togaviridae, Herpesviridae, Flaviviverdae, Rhabdoviridae, and Coronaviridae. Moreover, we show that Irgm1 knockout mice are highly resistant to chikungunya virus (CHIKV) infection. Altogether, our work highlights IRGM as a broad therapeutic target to promote defense against a large number of human viruses, including SARS-CoV-2, CHIKV, and Zika virus.


Subject(s)
GTP-Binding Proteins/antagonists & inhibitors , Virus Diseases/immunology , Animals , Antiviral Agents/pharmacology , Humans , Mice , Virus Replication
17.
Neuroscientist ; 28(5): 438-452, 2022 10.
Article in English | MEDLINE | ID: covidwho-1194426

ABSTRACT

The interactions of viruses with the nervous system were thought to be well understood until the recent outbreaks of Zika and SARS-CoV-2. In this review, we consider these emerging pathogens, the range and mechanisms of the neurological disease in humans, and how the biomedical research enterprise has pivoted to answer questions about viral pathogenesis, immune response, and the special vulnerability of the nervous system. ZIKV stands out as the only new virus in a generation, associating with congenital brain defects, neurological manifestations of microcephaly in newborns, and radiculopathy in adults. COVID-19, the disease caused by SARS-CoV-2, has swept the planet in an unprecedented manner and is feared worldwide for its effect on the respiratory system, but recent evidence points to important neurological sequelae. These can include anosmia, vasculopathy, paresthesias, and stroke. Evidence of ZIKV and SARS-CoV-2 genetic material from neural tissue, and evidence of infection of neural cells, raises questions about how these emerging viruses produce disease, and where new therapies might emerge.


Subject(s)
COVID-19 , Nervous System Diseases , Zika Virus Infection , Zika Virus , Adult , Humans , Infant, Newborn , SARS-CoV-2 , Zika Virus/genetics , Zika Virus Infection/complications , Zika Virus Infection/epidemiology
18.
Viruses ; 13(2)2021 01 28.
Article in English | MEDLINE | ID: covidwho-1060540

ABSTRACT

There have been reports of neurological abnormalities associated with the Zika virus (ZIKV), such as congenital Zika syndrome (CZS) in children born to mothers infected during pregnancy. We investigated how the immune response to ZIKV during pregnancy is primed and conduct a thorough evaluation of the inflammatory and cytotoxic profiles as well as the expression of CCR5 and CX3CR1. We compared the reactivity of T cells to ZIKV peptides in convalescent mothers infected during pregnancy. The child's clinical outcome (i.e., born with or without CZS) was taken to be the variable. The cells were stimulated in vitro with ZIKV peptides and evaluated using the ELISPOT and flow cytometry assays. After in vitro stimulation with ZIKV peptides, we observed a tendency toward a higher Interferon gamma (IFN-γ)-producing T cell responses in mothers who had asymptomatic children and a higher CD107a expression in T cells in mothers who had children with CZS. We found a higher frequency of T cells expressing CD107a+ and co-expressing CX3CR1+CCR5+, which is much clearer in the T cells of mothers who had CZS children. We suggest that this differential profile influenced the clinical outcome of babies. These data need to be further investigated, including the evaluation of other ZIKV peptides and markers and functional assays.


Subject(s)
CX3C Chemokine Receptor 1/metabolism , Pregnancy Complications, Infectious/immunology , Receptors, CCR5/metabolism , T-Lymphocytes/immunology , Zika Virus Infection/immunology , Adult , Cross-Sectional Studies , Cytotoxicity, Immunologic , Female , Humans , Infant , Interferon-gamma/metabolism , Lysosome-Associated Membrane Glycoproteins/metabolism , Pregnancy , Pregnancy Outcome , T-Lymphocytes/metabolism , Young Adult , Zika Virus/immunology
19.
Pathogens ; 10(1)2021 Jan 14.
Article in English | MEDLINE | ID: covidwho-1034743

ABSTRACT

In silico analysis is a promising approach for understanding biological events in complex diseases. Herein we report on the innovative computational workflow allowed to highlight new direct interactions between human transcription factors (TFs) and an entire genome of virus ZikaSPH2015 strain in order to identify the occurrence of specific motifs on a genomic Zika Virus sequence that is able to bind and, therefore, sequester host's TFs. The analysis pipeline was performed using different bioinformatics tools available online (free of charge). According to obtained results of this in silico analysis, it is possible to hypothesize that these TFs binding motifs might be able to explain the complex and heterogeneous phenotype presentation in Zika-virus-affected fetuses/newborns, as well as the less severe condition in adults. Moreover, the proposed in silico protocol identified thirty-three different TFs identical to the distribution of TFBSs (Transcription Factor Binding Sites) on ZikaSPH2015 strain, potentially able to influence genes and pathways with biological functions confirming that this approach could find potential answers on disease pathogenesis.

SELECTION OF CITATIONS
SEARCH DETAIL