Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.089
Filter
1.
PLoS One ; 19(8): e0307902, 2024.
Article in English | MEDLINE | ID: mdl-39116118

ABSTRACT

Microcephaly, Guillain-Barré syndrome, and potential sexual transmission stand as prominent complications associated with Zika virus (ZIKV) infection. The absence of FDA-approved drugs or vaccines presents a substantial obstacle in combatting the virus. Furthermore, the inclusion of pregnancy in the pharmacological screening process complicates and extends the endeavor to ensure molecular safety and minimal toxicity. Given its pivotal role in viral assembly and maturation, the NS2B-NS3 viral protease emerges as a promising therapeutic target against ZIKV. In this context, a dipeptide inhibitor was specifically chosen as a control against 200 compounds for docking analysis. Subsequent molecular dynamics simulations extending over 200 ns were conducted to ascertain the stability of the docked complex and confirm the binding of the inhibitor at the protein's active site. The simulation outcomes exhibited conformity to acceptable thresholds, encompassing parameters such as root mean square deviation (RMSD), root mean square fluctuation (RMSF), ligand-protein interaction analysis, ligand characterization, and surface area analysis. Notably, analysis of ligand angles bolstered the identification of prospective ligands capable of inhibiting viral protein activity and impeding virus dissemination. In this study, the integration of molecular docking and dynamics simulations has pinpointed the dipeptide inhibitor as a potential candidate ligand against ZIKV protease, thereby offering promise for therapeutic intervention against the virus.


Subject(s)
Dipeptides , Molecular Docking Simulation , Molecular Dynamics Simulation , Protease Inhibitors , Viral Nonstructural Proteins , Zika Virus , Zika Virus/enzymology , Zika Virus/drug effects , Dipeptides/chemistry , Dipeptides/pharmacology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Humans , Protein Binding , Viral Proteases , Nucleoside-Triphosphatase , DEAD-box RNA Helicases
2.
Sci Immunol ; 9(98): eadk9872, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39121194

ABSTRACT

The Aedes aegypti mosquito is a vector of many infectious agents, including flaviviruses such as Zika virus. Components of mosquito saliva have pleomorphic effects on the vertebrate host to enhance blood feeding, and these changes also create a favorable niche for pathogen replication and dissemination. Here, we demonstrate that human CD47, which is known to be involved in various immune processes, interacts with a 34-kilodalton mosquito salivary protein named Nest1. Nest1 is up-regulated in blood-fed female A. aegypti and facilitates Zika virus dissemination in human skin explants. Nest1 has a stronger affinity for CD47 than its natural ligand, signal regulatory protein α, competing for binding at the same interface. The interaction between Nest1 with CD47 suppresses phagocytosis by human macrophages and inhibits proinflammatory responses by white blood cells, thereby suppressing antiviral responses in the skin. This interaction elucidates how an arthropod protein alters the human response to promote arbovirus infectivity.


Subject(s)
Aedes , Skin , Zika Virus , Aedes/immunology , Aedes/virology , Animals , Humans , Skin/immunology , Skin/virology , Zika Virus/immunology , Zika Virus/physiology , Female , Insect Proteins/immunology , Zika Virus Infection/immunology , Salivary Proteins and Peptides/immunology , Mosquito Vectors/immunology , Mosquito Vectors/virology , CD47 Antigen
3.
Sci Rep ; 14(1): 18002, 2024 08 03.
Article in English | MEDLINE | ID: mdl-39097642

ABSTRACT

Zika virus (ZIKV) infection was first reported in 2015 in Brazil as causing microcephaly and other developmental abnormalities in newborns, leading to the identification of Congenital Zika Syndrome (CZS). Viral infections have been considered an environmental risk factor for neurodevelopmental disorders outcome, such as Autism Spectrum Disorder (ASD). Moreover, not only the infection per se, but maternal immune system activation during pregnancy, has been linked to fetal neurodevelopmental disorders. To understand the impact of ZIKV vertical infection on brain development, we derived induced pluripotent stem cells (iPSC) from Brazilian children born with CZS, some of the patients also being diagnosed with ASD. Comparing iPSC-derived neurons from CZS with a control group, we found lower levels of pre- and postsynaptic proteins and reduced functional synapses by puncta co-localization. Furthermore, neurons and astrocytes derived from the CZS group showed decreased glutamate levels. Additionally, the CZS group exhibited elevated levels of cytokine production, one of which being IL-6, already associated with the ASD phenotype. These preliminary findings suggest that ZIKV vertical infection may cause long-lasting disruptions in brain development during fetal stages, even in the absence of the virus after birth. These disruptions could contribute to neurodevelopmental disorders manifestations such as ASD. Our study contributes with novel knowledge of the CZS outcomes and paves the way for clinical validation and the development of potential interventions to mitigate the impact of ZIKV vertical infection on neurodevelopment.


Subject(s)
Brain , Induced Pluripotent Stem Cells , Infectious Disease Transmission, Vertical , Synapses , Zika Virus Infection , Zika Virus , Humans , Zika Virus Infection/virology , Zika Virus Infection/pathology , Female , Zika Virus/pathogenicity , Synapses/pathology , Synapses/metabolism , Brain/virology , Brain/pathology , Pregnancy , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/virology , Neurons/virology , Neurons/metabolism , Neurons/pathology , Male , Astrocytes/virology , Astrocytes/metabolism , Neuroinflammatory Diseases/virology , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/metabolism , Pregnancy Complications, Infectious/virology , Pregnancy Complications, Infectious/pathology , Brazil , Infant, Newborn , Autism Spectrum Disorder/virology , Child
4.
Mol Biomed ; 5(1): 30, 2024 08 03.
Article in English | MEDLINE | ID: mdl-39095588

ABSTRACT

Zika virus, a mosquito-borne arbovirus, has repeatedly caused large pandemics with symptoms worsening from mild and self-limiting diseases to Guillain-Barré syndrome in adults and fetal microcephaly in newborns. In recent years, Zika virus diseases have posed a serious threat to human health. The shortage of susceptible small animal models makes it difficult to study pathogenic mechanisms and evaluate potential therapies for Zika virus infection. Therefore, we chose immunocompromised mice (AG129 mice) deficient in IFN-α/ß and IFN-γ receptors, which can abolish the innate immune system that prevents Zika virus infection early. AG129 mice were infected with the Zika virus, and this mouse model exhibited replication dynamics, tissue tropism, pathological lesion and immune activation of the Zika virus. Our results suggest that the inoculum dose of Zika virus can affect the viral replication dynamics, cytokine responses and survival rate in AG129 mice. By testing the potential antiviral drug favipiravir, several critical indicators, including replication dynamics and survival rates, were identified in AG129 mice after Zika virus infection. It is suggested that the model is reliable for drug evaluation. In brief, this model provides a potential platform for studies of the infectivity, virulence, and pathogenesis of the Zika virus. Moreover, the development of an accessible mouse model of Zika virus infection will expedite the research and deployment of therapeutics and vaccines.


Subject(s)
Cytokines , Disease Models, Animal , Immunocompromised Host , Virus Replication , Zika Virus Infection , Zika Virus , Animals , Zika Virus/immunology , Zika Virus/pathogenicity , Zika Virus Infection/immunology , Zika Virus Infection/virology , Virus Replication/drug effects , Mice , Cytokines/metabolism , Survival Rate , Receptor, Interferon alpha-beta/genetics , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Interferon gamma Receptor , Vero Cells
5.
Sci Rep ; 14(1): 18112, 2024 08 05.
Article in English | MEDLINE | ID: mdl-39103482

ABSTRACT

This study presents a computational investigation of a stochastic Zika virus along with optimal control model using the Legendre spectral collocation method (LSCM). By accumulation of stochasticity into the model through the proposed stochastic differential equations, we appropriating the random fluctuations essential in the progression and disease transmission. The stability, convergence and accuracy properties of the LSCM are conscientiously analyzed and also demonstrating its strength for solving the complex epidemiological models. Moreover, the study evaluates the various control strategies, such as treatment, prevention and treatment pesticide control, and identifies optimal combinations that the intervention costs and also minimize the proposed infection rates. The basic properties of the given model, such as the reproduction number, were determined with and without the presence of the control strategies. For R 0 < 0 , the model satisfies the disease-free equilibrium, in this case the disease die out after some time, while for R 0 > 1 , then endemic equilibrium is satisfied, in this case the disease spread in the population at higher scale. The fundamental findings acknowledge the significant impact of stochastic phonemes on the robustness and effectiveness of control strategies that accelerating the need for cost-effective and multi-faceted approaches. In last the results provide the valuable insights for public health department to enabling more impressive mitigation of Zika virus outbreaks and management in real-world scenarios.


Subject(s)
Stochastic Processes , Zika Virus Infection , Zika Virus , Zika Virus Infection/epidemiology , Zika Virus Infection/prevention & control , Zika Virus Infection/transmission , Humans , Zika Virus/physiology , Computer Simulation , Epidemiological Models
6.
Proc Natl Acad Sci U S A ; 121(34): e2403235121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39145933

ABSTRACT

The ZIKA virus (ZIKV) evades the host immune response by degrading STAT2 through its NS5 protein, thereby inhibiting type I interferon (IFN)-mediated antiviral immunity. However, the molecular mechanism underlying this process has remained elusive. In this study, we performed a genome-wide CRISPR/Cas9 screen, revealing that ZSWIM8 as the substrate receptor of Cullin3-RING E3 ligase is required for NS5-mediated STAT2 degradation. Genetic depletion of ZSWIM8 and CUL3 substantially impeded NS5-mediated STAT2 degradation. Biochemical analysis illuminated that NS5 enhances the interaction between STAT2 and the ZSWIM8-CUL3 E3 ligase complex, thereby facilitating STAT2 ubiquitination. Moreover, ZSWIM8 knockout endowed A549 and Huh7 cells with partial resistance to ZIKV infection and protected cells from the cytopathic effects induced by ZIKV, which was attributed to the restoration of STAT2 levels and the activation of IFN signaling. Subsequent studies in a physiologically relevant model, utilizing human neural progenitor cells, demonstrated that ZSWIM8 depletion reduced ZIKV infection, resulting from enhanced IFN signaling attributed to the sustained levels of STAT2. Our findings shed light on the role of ZIKV NS5, serving as the scaffold protein, reprograms the ZSWIM8-CUL3 E3 ligase complex to orchestrate STAT2 proteasome-dependent degradation, thereby facilitating evasion of IFN antiviral signaling. Our study provides unique insights into ZIKV-host interactions and holds promise for the development of antivirals and prophylactic vaccines.


Subject(s)
Cullin Proteins , Interferon Type I , Proteolysis , STAT2 Transcription Factor , Signal Transduction , Ubiquitin-Protein Ligases , Ubiquitination , Viral Nonstructural Proteins , Zika Virus Infection , Zika Virus , Humans , STAT2 Transcription Factor/metabolism , Zika Virus/immunology , Zika Virus/physiology , Zika Virus/metabolism , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics , Interferon Type I/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Zika Virus Infection/metabolism , Zika Virus Infection/immunology , Zika Virus Infection/virology , Cullin Proteins/metabolism , A549 Cells , HEK293 Cells , CRISPR-Cas Systems
7.
Sci Rep ; 14(1): 18470, 2024 08 09.
Article in English | MEDLINE | ID: mdl-39122799

ABSTRACT

The microbial communities residing in the mosquito midgut play a key role in determining the outcome of mosquito pathogen infection. Elizabethkingia anophelis, originally isolated from the midgut of Anopheles gambiae possess a broad-spectrum antiviral phenotype, yet a gap in knowledge regarding the mechanistic basis of its interaction with viruses exists. The current study aims to identify pathways and genetic factors linked to E. anophelis antiviral activity. The understanding of E. anophelis antiviral mechanism could lead to novel transmission barrier tools to prevent arboviral outbreaks. We utilized a non-targeted multi-omics approach, analyzing extracellular lipids, proteins, metabolites of culture supernatants coinfected with ZIKV and E. anophelis. We observed a significant decrease in arginine and phenylalanine levels, metabolites that are essential for viral replication and progression of viral infection. This study provides insights into the molecular basis of E. anophelis antiviral phenotype. The findings lay a foundation for in-depth mechanistic studies.


Subject(s)
Flavobacteriaceae , Zika Virus , Zika Virus/physiology , Animals , Flavobacteriaceae/metabolism , Flavobacteriaceae/genetics , Anopheles/virology , Anopheles/microbiology , Zika Virus Infection/virology , Antiviral Agents/pharmacology , Antiviral Agents/metabolism , Virus Replication , Phenylalanine/metabolism , Arginine/metabolism , Multiomics
8.
Virus Res ; 348: 199447, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39117146

ABSTRACT

One third of all emerging infectious diseases are vector-borne, with no licensed antiviral therapies available against any vector-borne viruses. Zika virus and Usutu virus are two emerging flaviviruses transmitted primarily by mosquitoes. These viruses modulate different host pathways, including the PI3K/AKT/mTOR pathway. Here, we report the effect on ZIKV and USUV replication of two AKT inhibitors, Miransertib (ARQ-092, allosteric inhibitor) and Capivasertib (AZD5363, competitive inhibitor) in different mammalian and mosquito cell lines. Miransertib showed a stronger inhibitory effect against ZIKV and USUV than Capivasertib in mammalian cells, while Capivasertib showed a stronger effect in mosquito cells. These findings indicate that AKT plays a conserved role in flavivirus infection, in both the vertebrate host and invertebrate vector. Nevertheless, the specific function of AKT may vary depending on the host species. These findings indicate that AKT may be playing a conserved role in flavivirus infection in both, the vertebrate host and the invertebrate vector. However, the specific function of AKT may vary depending on the host species. A better understanding of virus-host interactions is therefore required to develop new treatments to prevent human disease and new approaches to control transmission by insect vectors.


Subject(s)
Flavivirus Infections , Flavivirus , Proto-Oncogene Proteins c-akt , Virus Replication , Zika Virus , Animals , Flavivirus/physiology , Flavivirus/drug effects , Flavivirus/genetics , Humans , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Cell Line , Zika Virus/physiology , Zika Virus/drug effects , Flavivirus Infections/virology , Flavivirus Infections/transmission , Vertebrates/virology , Antiviral Agents/pharmacology , Mosquito Vectors/virology , Chlorocebus aethiops , Culicidae/virology , Host-Pathogen Interactions
9.
RNA Biol ; 21(1): 1-10, 2024 Jan.
Article in English | MEDLINE | ID: mdl-39183472

ABSTRACT

One of the most recent advances in the analysis of viral RNA-cellular protein interactions is the Comprehensive Identification of RNA-binding Proteins by Mass Spectrometry (ChIRP-MS). Here, we used ChIRP-MS in mock-infected and Zika-infected wild-type cells and cells knockout for the zinc finger CCCH-type antiviral protein 1 (ZAP). We characterized 'ZAP-independent' and 'ZAP-dependent' cellular protein interactomes associated with flavivirus RNA and found that ZAP affects cellular proteins associated with Zika virus RNA. The ZAP-dependent interactome identified with ChIRP-MS provides potential ZAP co-factors for antiviral activity against Zika virus and possibly other viruses. Identifying the full spectrum of ZAP co-factors and mechanisms of how they act will be critical to understanding the ZAP antiviral system and may contribute to the development of antivirals.


Subject(s)
RNA, Viral , RNA-Binding Proteins , Zika Virus Infection , Zika Virus , Zika Virus/genetics , Zika Virus/physiology , Zika Virus/metabolism , Humans , RNA, Viral/metabolism , RNA, Viral/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Zika Virus Infection/virology , Zika Virus Infection/metabolism , Protein Binding , Host-Pathogen Interactions/genetics , Mass Spectrometry , HEK293 Cells
10.
Indian J Public Health ; 68(2): 163-166, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38953800

ABSTRACT

BACKGROUND: Several sporadic cases and outbreaks of Zika virus disease have been reported from different states of India. OBJECTIVES: This paper explored the possibility of any ongoing transmission of Zika virus (ZIKV) in the Bhopal region of Central India, where the last outbreak of this disease was reported in 2018. MATERIALS AND METHODS: We screened a group of 75 febrile patients who had already tested negative for the locally endemic causes of fever like dengue, chikungunya, enteric fever, malaria, and scrub typhus and two groups of asymptomatic healthy individuals represented by blood donors (n = 75) and antenatal mothers (n = 75). We tested blood samples of febrile patients for ZIKV RNA using real-time polymerase chain reaction (PCR), and for the healthy individuals, we determined anti-zika immunoglobulin G (IgG) antibodies using enzyme-linked immunosorbent assay. RESULTS: ZIKV RNA was not detected in any of the 75 samples tested by real-time PCR assay. Among the voluntary blood donors and antenatal mothers, a total of 10 (15.38%) and 5 (6.66%) individuals were found to be seropositive for anti-ZIKV IgG antibodies, respectively. The seropositive group was found to have higher age 33.06 (±10.83) years as compared to seronegative individuals 26.60 (±5.12) years (P = 0.037). CONCLUSION: This study, which is the first survey of seroprevalence of anti-Zika antibodies from India, reports an overall seropositivity rate of 10% for anti-Zika antibodies among the healthy population, suggesting an ongoing, low level, silent transmission of ZIKV in the local community.


Subject(s)
Zika Virus Infection , Zika Virus , Humans , India/epidemiology , Zika Virus Infection/epidemiology , Zika Virus Infection/transmission , Seroepidemiologic Studies , Adult , Female , Pilot Projects , Male , Zika Virus/immunology , Zika Virus/isolation & purification , Immunoglobulin G/blood , Young Adult , Antibodies, Viral/blood , Middle Aged , RNA, Viral , Adolescent , Enzyme-Linked Immunosorbent Assay , Real-Time Polymerase Chain Reaction
11.
FASEB J ; 38(13): e23799, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38979938

ABSTRACT

Maternal Zika virus (ZIKV) infection during pregnancy has been associated with severe intrauterine growth restriction (IUGR), placental damage, metabolism disturbances, and newborn neurological abnormalities. Here, we investigated the impact of maternal ZIKV infection on placental nutrient transporters and nutrient-sensitive pathways. Immunocompetent (C57BL/6) mice were injected with Low (103 PFU-ZIKVPE243) or High (5 × 107 PFU-ZIKVPE243) ZIKV titers at gestational day (GD) 12.5, and tissue was collected at GD18.5 (term). Fetal-placental growth was impaired in male fetuses, which exhibited higher placental expression of the ZIKV infective marker, eukaryotic translation initiation factor 2 (eIF2α), but lower levels of phospho-eIF2α. There were no differences in fetal-placental growth in female fetuses, which exhibited no significant alterations in placental ZIKV infective markers. Furthermore, ZIKV promoted increased expression of glucose transporter type 1 (Slc2a1/Glut1) and decreased levels of glucose-6-phosphate in female placentae, with no differences in amino acid transport potential. In contrast, ZIKV did not impact glucose transporters in male placentae but downregulated sodium-coupled neutral amino acid 2 (Snat2) transporter expression. We also observed sex-dependent differences in the hexosamine biosynthesis pathway (HBP) and O-GlcNAcylation in ZIKV-infected pregnancies, showing that ZIKV can disturb placental nutrient sensing. Our findings highlight molecular alterations in the placenta caused by maternal ZIKV infection, shedding light on nutrient transport, sensing, and availability. Our results also suggest that female and male placentae employ distinct coping mechanisms in response to ZIKV-induced metabolic changes, providing insights into therapeutic approaches for congenital Zika syndrome.


Subject(s)
Fetal Development , Mice, Inbred C57BL , Placenta , Signal Transduction , Zika Virus Infection , Zika Virus , Animals , Female , Zika Virus Infection/metabolism , Zika Virus Infection/virology , Pregnancy , Mice , Placenta/metabolism , Placenta/virology , Male , Fetal Development/physiology , Pregnancy Complications, Infectious/virology , Pregnancy Complications, Infectious/metabolism , Nutrients/metabolism , Glucose Transporter Type 1/metabolism
12.
Front Cell Infect Microbiol ; 14: 1421744, 2024.
Article in English | MEDLINE | ID: mdl-38988809

ABSTRACT

The increase in incidence and geographical expansion of viruses transmitted by the Aedes mosquitoes, such as dengue (DENV) and zika (ZIKV) in the Americas, represents a burden for healthcare systems in tropical and subtropical regions. These and other under-detected arboviruses co-circulate in Costa Rica, adding additional complexity to their management due to their shared epidemiological behavior and similarity of symptoms in early stages. Since diagnostics of febrile illness is mostly based on clinical symptoms alone, we gathered acute-phase serum and urine from 399 samples of acute dengue-like cases from two healthcare facilities of Costa Rica, during an outbreak of arboviruses from July 2017 to May 2018, and tested them using molecular and serological methods. The analyses showed that of the clinically presumptive arbovirus cases that were reported, only 39.4% (n=153) of the samples were confirmed positive by RT-PCR to be DENV (DENV (10.3%), CHIKV (0.2%), ZIKV (27.3%), or mixed infections (1.5%). RT-PCR for other alphaviruses and flaviviruses, and PCR for Leptospira sp were negative. Furthermore, to assess flavivirus positivity in post-acute patients, the negative sera were tested against Dengue-IgM. 20% of sera were found positive, confounding even more the definitive number of cases, and emphasizing the need of several distinct diagnostic tools for accurate diagnostics. Molecular characterization of the prM and E genes from isolated viruses revealed that the American/Asian genotype of DENV-2 and the Asian lineage of ZIKV were circulating during this outbreak. Two different clades of DENV-2 American/Asian genotype were identified to co-circulate in the same region and a difference in the platelet and leukocyte count was noted between people infected with each clade, suggesting a putative distinct virulence. Our study sheds light on the necessity for healthcare strategies in managing arbovirus outbreaks, emphasizing the importance of comprehensive molecular and serological diagnostic approaches, as well as molecular characterization. This approach aids in enhancing our understanding of the clinical and epidemiological aspects of arboviral diseases during outbreaks. Our research highlights the need to strengthen training programs for health professionals and the need to increase research-based on laboratory evidence for diagnostic accuracy, guidance, development and implementation of public health interventions and epidemiological surveillance.


Subject(s)
Dengue Virus , Dengue , Disease Outbreaks , Zika Virus Infection , Zika Virus , Humans , Costa Rica/epidemiology , Dengue/epidemiology , Dengue/diagnosis , Dengue/virology , Zika Virus Infection/epidemiology , Zika Virus Infection/diagnosis , Zika Virus Infection/virology , Zika Virus/genetics , Zika Virus/isolation & purification , Dengue Virus/genetics , Dengue Virus/isolation & purification , Dengue Virus/classification , Female , Male , Adult , Adolescent , Middle Aged , Young Adult , Child , Child, Preschool , Aged , Caribbean Region/epidemiology , Phylogeny , Infant , Animals , Coinfection/epidemiology , Coinfection/virology , Aged, 80 and over , Antibodies, Viral/blood
13.
PLoS Negl Trop Dis ; 18(7): e0012066, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38968296

ABSTRACT

Zika virus (ZIKV) has become a global health problem over the past decade due to the extension of the geographic distribution of the Asian/American genotype. Recent epidemics of Asian/American ZIKV have been associated with developmental disorders in humans. There is mounting evidence that African ZIKV may be associated with increased fetal pathogenicity necessitating to pay a greater attention towards currently circulating viral strains in sub-Saharan Africa. Here, we generated an infectious molecular clone GUINEA-18 of a recently transmitted human ZIKV isolate from West Africa, ZIKV-15555. The available infectious molecular clone MR766MC of historical African ZIKV strain MR766-NIID was used for a molecular clone-based comparative study. Viral clones GUINEA-18 and MR766MC were compared for their ability to replicate in VeroE6, A549 and HCM3 cell lines. There was a lower replication rate for GUINEA-18 associated with weaker cytotoxicity and reduced innate immune system activation compared with MR766MC. Analysis of chimeric viruses between viral clones stressed the importance of NS1 to NS4B proteins, with a particular focus of NS4B on GUINEA-18 replicative properties. ZIKV has developed strategies to prevent cytoplasmic stress granule formation which occurs in response to virus infection. GUINEA-18 was greatly efficient in inhibiting stress granule assembly in A549 cells subjected to a physiological stressor, with NS1 to NS4B proteins also being critical in this process. The impact of these GUINEA-18 proteins on viral replicative abilities and host-cell responses to viral infection raises the question of the role of nonstructural proteins in the pathogenicity of currently circulating ZIKV in sub-Saharan Africa.


Subject(s)
Virus Replication , Zika Virus Infection , Zika Virus , Zika Virus/genetics , Zika Virus/physiology , Humans , Africa, Western/epidemiology , Zika Virus Infection/virology , Animals , Chlorocebus aethiops , Cell Line , Vero Cells , A549 Cells
14.
PLoS Pathog ; 20(7): e1012408, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39038037

ABSTRACT

c-FLIP functions as a dual regulator of apoptosis and inflammation, yet its implications in Zika virus (ZIKV) infection remain partially understood, especially in the context of ZIKV-induced congenital Zika syndrome (CZS) where both apoptosis and inflammation play pivotal roles. Our findings demonstrate that c-FLIP promotes ZIKV infection in placental cells and myeloid-derived macrophages, involving inflammation and caspase-8/3-mediated apoptosis. Moreover, our observations reveal that c-FLIP augments ZIKV infection in multiple tissues, including blood cell, spleen, uterus, testis, and the brain of mice. Notably, the partial deficiency of c-FLIP provides protection to embryos against ZIKV-induced CZS, accompanied by a reduction in caspase-3-mediated apoptosis. Additionally, we have found a distinctive parental effect of c-FLIP influencing ZIKV replication in fetal heads. In summary, our study reveals the critical role of c-FLIP as a positive regulator in caspase-8/3-mediated apoptosis during ZIKV infection, significantly contributing to the development of CZS.


Subject(s)
Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein , Caspase 3 , Caspase 8 , Zika Virus Infection , Zika Virus , Zika Virus Infection/virology , Zika Virus Infection/metabolism , Zika Virus Infection/pathology , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Animals , Mice , Caspase 8/metabolism , Female , Humans , Caspase 3/metabolism , Pregnancy , Placenta/virology , Placenta/metabolism , Placenta/pathology , Mice, Inbred C57BL , Virus Replication , Mice, Knockout
15.
Viruses ; 16(7)2024 Jun 22.
Article in English | MEDLINE | ID: mdl-39066166

ABSTRACT

AIM: Congenital Zika Virus Syndrome (CZS) presents notable hurdles to neurodevelopment, with language development emerging as a crucial aspect. This study investigates sleep patterns and language skills in children with CZS, aiming to explore the potential synchronization of sleep development with their neurodevelopment. METHOD: We studied cross-sectionally 135 children with CZS aged 0 to 48 months, investigating sleep using the BISQ Questionnaire. Language development was assessed using the Early Language Milestone Scale, while motor development and cognitive and social ability were assessed using the Bayley Scales of Infant and Young Child Development 3rd edition. We also studied longitudinally a cohort of 16 children (initially aged 0 to 12 months) whom we followed for four years, assessing at one-year intervals. RESULTS: Sleep disturbances and language deficits were highly frequent in this population. In the 0-12 months group, a late bedtime and frequent nighttime awakenings were associated with poorer auditory expressive skills. At 13-24 months, nighttime awakenings were associated with poorer auditory expressive skills, while among 25-36-month-olds decreased auditory receptive skills were associated with longer sleep onset latency and reduced nighttime sleep duration. CONCLUSION: The brain alterations caused by Zika virus infection affect both sleep disturbances and delays in language development. It is possible that sleep disturbance may be a mediating factor in the pathway between CZS and delayed language development, as the three analyzed language skills showed a correlation with sleep parameters.


Subject(s)
Language Development , Sleep , Zika Virus Infection , Humans , Zika Virus Infection/complications , Zika Virus Infection/physiopathology , Zika Virus Infection/virology , Zika Virus Infection/congenital , Infant , Female , Male , Child, Preschool , Sleep/physiology , Infant, Newborn , Cross-Sectional Studies , Sleep Wake Disorders/physiopathology , Sleep Wake Disorders/virology , Zika Virus/physiology , Longitudinal Studies , Surveys and Questionnaires , Language Development Disorders/physiopathology , Language Development Disorders/virology
16.
Viruses ; 16(7)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39066208

ABSTRACT

Little is known about the frequency of Zika virus (ZIKV) infections in Sudan. The aim of this study was to obtain data on the prevalence of ZIKV infections and the immunity of the population in the country. To this end, 198 sera obtained between December 2012 and January 2013 in different regions in Sudan were examined for neutralizing antibodies against ZIKV, dengue virus (DENV), and yellow fever virus (YFV). The sera were non-randomly selected. The neutralization titers were compared with each other and with the WHO 1st International Standard for anti-Asian lineage Zika virus antibody. Twenty-six sera neutralized ZIKV. One-third of these sera had higher neutralization titers against ZIKV than against DENV-2 and -3. Two sera showed higher neutralization titers than the WHO standard for ZIKV antibodies. These data suggest occasional ZIKV infections in Sudan. The low percentage of sera in this cohort that neutralized ZIKV indicates that, in the study period, the population was susceptible to ZIKV infection.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Zika Virus Infection , Zika Virus , Sudan/epidemiology , Humans , Zika Virus Infection/epidemiology , Zika Virus Infection/immunology , Zika Virus Infection/blood , Antibodies, Viral/blood , Zika Virus/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Adult , Female , Male , Middle Aged , Young Adult , Adolescent , Neutralization Tests , Dengue Virus/immunology , Child , Yellow fever virus/immunology , Seroepidemiologic Studies , Child, Preschool , Aged , Prevalence
17.
Acta Trop ; 257: 107321, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38972559

ABSTRACT

Fragmented landscapes in Mexico, characterized by a mix of agricultural, urban, and native vegetation cover, presents unique ecological characteristics that shape the mosquito community composition and mosquito-borne diseases. The extent to which landscape influences mosquito populations and mosquito-borne diseases is still poorly understood. This work assessed the effect of landscape metrics -agriculture, urban, and native vegetation cover- on mosquito diversity and arbovirus presence in fragmented tropical deciduous forests in Central Mexico during 2021. Among the 21 mosquito species across six genera we identified, Culex quinquefasciatus was the most prevalent species, followed by Aedes aegypti, Ae. albopictus, and Ae. epactius. Notably, areas with denser native vegetation cover displayed higher mosquito species richness, which could have an impact on phenomena such as the dilution effect. Zika and dengue virus were detected in 85% of captured species, with first reports of DENV in several Aedes species and ZIKV in multiple Aedes and Culex species. These findings underscore the necessity of expanding arbovirus surveillance beyond Ae. aegypti and advocate for a deeper understanding of vector ecology in fragmented landscapes to adequately address public health strategies.


Subject(s)
Arboviruses , Biodiversity , Culicidae , Mosquito Vectors , Animals , Arboviruses/isolation & purification , Arboviruses/classification , Mexico/epidemiology , Mosquito Vectors/virology , Mosquito Vectors/classification , Culicidae/virology , Culicidae/classification , Agriculture , Aedes/virology , Aedes/classification , Cities , Zika Virus/isolation & purification , Zika Virus/genetics , Ecosystem
18.
Molecules ; 29(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39064900

ABSTRACT

Pathogenic microorganisms play a crucial role in the global disease burden due to their ability to cause various diseases and spread through multiple transmission routes. Immunity tests identify antigens related to these pathogens, thereby confirming past infections and monitoring the host's immune response. Traditional pathogen detection methods, including enzyme-linked immunosorbent assays (ELISAs) and chemiluminescent immunoassays (CLIAs), are often labor-intensive, slow, and reliant on sophisticated equipment and skilled personnel, which can be limiting in resource-poor settings. In contrast, the development of microfluidic technologies presents a promising alternative, offering automation, miniaturization, and cost efficiency. These advanced methods are poised to replace traditional assays by streamlining processes and enabling rapid, high-throughput immunity testing for pathogens. This review highlights the latest advancements in microfluidic systems designed for rapid and high-throughput immunity testing, incorporating immunosensors, single molecule arrays (Simoas), a lateral flow assay (LFA), and smartphone integration. It focuses on key pathogenic microorganisms such as SARS-CoV-2, influenza, and the ZIKA virus (ZIKV). Additionally, the review discusses the challenges, commercialization prospects, and future directions to advance microfluidic systems for infectious disease detection.


Subject(s)
SARS-CoV-2 , Humans , SARS-CoV-2/immunology , SARS-CoV-2/isolation & purification , Microfluidics/methods , Microfluidics/instrumentation , COVID-19/immunology , COVID-19/diagnosis , COVID-19/virology , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Immunoassay/methods , Zika Virus/immunology , Lab-On-A-Chip Devices , Biosensing Techniques/methods , Influenza, Human/diagnosis , Influenza, Human/immunology , Zika Virus Infection/diagnosis , Zika Virus Infection/immunology
19.
Virologie (Montrouge) ; 28(3): 187-197, 2024 Jun 01.
Article in French | MEDLINE | ID: mdl-38970340

ABSTRACT

Orthoflaviviruses are enveloped positive-sense RNA viruses comprising numerous human pathogens transmitted by hematophagous arthropods. This includes viruses such as dengue virus, Zika virus, and yellow fever virus. The viral nonstructural protein NS1 plays a central role in the pathogenesis and cycle of these viruses by acting in two different forms: associated with the plasma membrane (NS1m) or secreted outside the cell (NS1s). The versatility of NS1 is evident in its ability to modulate various aspects of the infectious process, from immune evasion to pathogenesis. As an intracellular protein, it disrupts many processes, interfering with signaling pathways and facilitating viral replication in concert with other viral proteins. As a secreted protein, NS1 actively participates in immune evasion, interfering with the host immune system, inhibiting the complement system, facilitating viral dissemination, and disrupting the integrity of endothelial barriers. This review primarily aims to address the role of NS1 in viral pathogenesis associated with orthoflaviviruses.


Subject(s)
Viral Nonstructural Proteins , Virus Replication , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/physiology , Humans , Animals , Flavivirus Infections/virology , Immune Evasion , Flavivirus/physiology , Flavivirus/pathogenicity , Zika Virus/physiology , Zika Virus/pathogenicity , Dengue Virus/physiology
20.
J Pregnancy ; 2024: 1758662, 2024.
Article in English | MEDLINE | ID: mdl-38961858

ABSTRACT

Congenital Zika syndrome (CZS) is a major concern in India and highlights the multifaceted challenges posed by the Zika virus (ZIKV). The alarming increase in CZS cases in India, a condition that has serious effects on both public health and newborns, has raised concerns. This review highlights the importance of raising concern and awareness and taking preventive measures by studying the epidemiology, clinical symptoms, and potential long-term consequences of CZS. The review also contributes to worldwide research and information sharing to improve the understanding and prevention of CZS. As India deals with the changing nature of CZS, this thorough review is an important tool for policymakers, health workers, and researchers to understand what is happening now, plan for what to do in the future, and work together as a team, using medical knowledge, community involvement, and study projects to protect newborns' health and reduce the public health impact of these syndromes.


Subject(s)
Pregnancy Complications, Infectious , Zika Virus Infection , Humans , Zika Virus Infection/epidemiology , Zika Virus Infection/congenital , Zika Virus Infection/prevention & control , Zika Virus Infection/complications , India/epidemiology , Pregnancy , Infant, Newborn , Pregnancy Complications, Infectious/epidemiology , Pregnancy Complications, Infectious/prevention & control , Female , Zika Virus , Microcephaly/epidemiology , Microcephaly/virology , Microcephaly/etiology
SELECTION OF CITATIONS
SEARCH DETAIL