Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
PLoS Negl Trop Dis ; 17(2): e0011095, 2023 02.
Article in English | MEDLINE | ID: mdl-36735632

ABSTRACT

Mosquito saliva is a mix of numerous proteins that are injected into the skin while the mosquito searches for a blood meal. While mosquito saliva is known to be immunogenic, the salivary components driving these immune responses, as well as the types of immune responses that occur, are not well characterized. We investigated the effects of one potential immunomodulatory mosquito saliva protein, sialokinin, on the human immune response. We used flow cytometry to compare human immune cell populations between humanized mice bitten by sialokinin knockout mosquitoes or injected with sialokinin, and compared them to those bitten by wild-type mosquitoes, unbitten, or saline-injected control mice. Humanized mice received 4 mosquito bites or a single injection, were euthanized after 7 days, and skin, spleen, bone marrow, and blood were harvested for immune cell profiling. Our results show that bites from sialokinin knockout mosquitoes induced monocyte and macrophage populations in the skin, blood, bone marrow, and spleens, and primarily affected CD11c- cell populations. Other increased immune cells included plasmacytoid dendritic cells in the blood, natural killer cells in the skin and blood, and CD4+ T cells in all samples analyzed. Conversely, we observed that mice bitten with sialokinin knockout mosquitoes had decreased NKT cell populations in the skin, and fewer B cells in the blood, spleen, and bone marrow. Taken together, we demonstrated that sialokinin knockout saliva induces elements of a TH1 cellular immune response, suggesting that the sialokinin peptide is inducing a TH2 cellular immune response during wild-type mosquito biting. These findings are an important step towards understanding how mosquito saliva modulates the human immune system and which components of saliva may be critical for arboviral infection. By identifying immunomodulatory salivary proteins, such as sialokinin, we can develop vaccines against mosquito saliva components and direct efforts towards blocking arboviral infections.


Subject(s)
Aedes , Saliva , Humans , Animals , Mice , Spleen , Skin , Immunity , CD4-Positive T-Lymphocytes , Mosquito Vectors , Aedes/physiology
3.
Eur J Med Chem ; 225: 113767, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34450494

ABSTRACT

Zika virus belongs to the Flavivirus family of RNA viruses, which include other important human pathogens such as dengue and West Nile virus. There are no approved antiviral drugs for these viruses. The highly conserved NS2B-NS3 protease of Flavivirus is essential for the replication of these viruses and it is therefore a drug target. Compound screen followed by medicinal chemistry optimization yielded a novel series of 2,6-disubstituted indole compounds that are potent inhibitors of Zika virus protease (ZVpro) with IC50 values as low as 320 nM. The structure-activity relationships of these and related compounds are discussed. Enzyme kinetics studies show the inhibitor 66 most likely exhibited a non-competitive mode of inhibition. In addition, this series of ZVpro inhibitors also inhibit the NS2B-NS3 protease of dengue and West Nile virus with reduced potencies. The most potent compounds 66 and 67 strongly inhibited Zika virus replication in cells with EC68 values of 1-3 µM. These compounds are novel pharmacological leads for further drug development targeting Zika virus.


Subject(s)
Antiviral Agents/pharmacology , Indoles/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Zika Virus/drug effects , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Indoles/chemical synthesis , Indoles/chemistry , Microbial Sensitivity Tests , Molecular Structure , RNA Helicases/antagonists & inhibitors , RNA Helicases/metabolism , Serine Endopeptidases/metabolism , Structure-Activity Relationship , Viral Nonstructural Proteins/metabolism
4.
PLoS Negl Trop Dis ; 15(6): e0009427, 2021 06.
Article in English | MEDLINE | ID: mdl-34106915

ABSTRACT

Chikungunya virus (CHIKV) is an emerging, mosquito-borne alphavirus responsible for acute to chronic arthralgias and neuropathies. Although it originated in central Africa, recent reports of disease have come from many parts of the world, including the Americas. While limiting human CHIKV cases through mosquito control has been used, it has not been entirely successful. There are currently no licensed vaccines or treatments specific for CHIKV disease, thus more work is needed to develop effective countermeasures. Current animal research on CHIKV is often not representative of human disease. Most models use CHIKV needle inoculation via unnatural routes to create immediate viremia and localized clinical signs; these methods neglect the natural route of transmission (the mosquito vector bite) and the associated human immune response. Since mosquito saliva has been shown to have a profound effect on viral pathogenesis, we evaluated a novel model of infection that included the natural vector, Aedes species mosquitoes, transmitting CHIKV to mice containing components of the human immune system. Humanized mice infected by 3-6 mosquito bites showed signs of systemic infection, with demonstrable viremia (by qRT-PCR and immunofluorescent antibody assay), mild to moderate clinical signs (by observation, histology, and immunohistochemistry), and immune responses consistent with human infection (by flow cytometry and IgM ELISA). This model should give a better understanding of human CHIKV disease and allow for more realistic evaluations of mechanisms of pathogenesis, prophylaxis, and treatments.


Subject(s)
Aedes/virology , Chikungunya Fever/pathology , Chikungunya Fever/transmission , Chikungunya virus/isolation & purification , Insect Bites and Stings , Animals , Antibodies, Monoclonal, Humanized , Antibodies, Viral/blood , Chlorocebus aethiops , Immunoglobulin M/blood , Mice , Mosquito Vectors , Needles , RNA, Viral/blood , Serologic Tests , Vero Cells
5.
J Med Chem ; 64(5): 2777-2800, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33596380

ABSTRACT

Flaviviruses, including Zika, dengue, and West Nile viruses, are important human pathogens. The highly conserved NS2B-NS3 protease of Flavivirus is essential for viral replication and therefore a promising drug target. Through compound screening, followed by medicinal chemistry studies, a novel series of 2,5,6-trisubstituted pyrazine compounds are found to be potent, allosteric inhibitors of Zika virus protease (ZVpro) with IC50 values as low as 130 nM. Their structure-activity relationships are discussed. The ZVpro inhibitors also inhibit homologous proteases of dengue and West Nile viruses, and their inhibitory activities are correlated. The most potent compounds 47 and 103 potently inhibited Zika virus replication in cells with EC68 values of 300-600 nM and in a mouse model of Zika infection. These compounds represent novel pharmacological leads for drug development against Flavivirus infections.


Subject(s)
Antiviral Agents/therapeutic use , Pyrazines/therapeutic use , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/therapeutic use , Viral Proteins/metabolism , Zika Virus Infection/drug therapy , Allosteric Regulation/drug effects , Animals , Antiviral Agents/chemical synthesis , Cell Line, Tumor , Dengue Virus/enzymology , Humans , Mice , Molecular Structure , Pyrazines/chemical synthesis , Serine Proteinase Inhibitors/chemical synthesis , Structure-Activity Relationship , Viral Nonstructural Proteins/antagonists & inhibitors , Virus Replication/drug effects , West Nile virus/enzymology , Zika Virus/enzymology
6.
PLoS One ; 15(12): e0244587, 2020.
Article in English | MEDLINE | ID: mdl-33378361

ABSTRACT

Our previous studies have shown that Zika virus (ZIKV) replicates in human prostate cells, suggesting that the prostate may serve as a long-term reservoir for virus transmission. Here, we demonstrated that the innate immune responses generated to three distinct ZIKV strains (all isolated from human serum) were significantly different and dependent on their passage history (in mosquito, monkey, or human cells). In addition, some of these phenotypic differences were reduced by a single additional cell culture passage, suggesting that viruses that have been passaged more than 3 times from the patient sample will no longer reflect natural phenotypes. Two of the ZIKV strains analyzed induced high levels of the IP-10 chemokine and IFNγ in human prostate epithelial and stromal mesenchymal stem cells. To further understand the importance of these innate responses on ZIKV replication, we measured the effects of IP-10 and its downstream receptor, CXCR3, on RNA and virus production in prostate cells. Treatment with IP-10, CXCR3 agonist, or CXCR3 antagonist significantly altered ZIKV viral gene expression, depending on their passage in cells of relevant hosts (mosquito or human). We detected differences in gene expression of two primary CXCR3 isoforms (CXCR3-A and CXCR3-B) on the two cell types, possibly explaining differences in viral output. Lastly, we examined the effects of IP-10, agonist, or antagonist on cell death and proliferation under physiologically relevant infection rates, and detected no significant differences. Although we did not measure protein expression directly, our results indicate that CXCR3 signaling may be a target for therapeutics, to ultimately stop sexual transmission of this virus.


Subject(s)
Chemokine CXCL10/metabolism , Prostate/virology , Receptors, CXCR3/metabolism , Zika Virus Infection/immunology , Zika Virus/physiology , Animals , Cell Line , Cell Proliferation , Cell Survival , Chemokine CXCL10/genetics , Culicidae/virology , Gene Expression Regulation , Haplorhini/virology , Humans , Immunity, Innate , Male , Prostate/cytology , Prostate/immunology , Receptors, CXCR3/genetics , Serial Passage , Signal Transduction , Virus Replication , Zika Virus/immunology , Zika Virus Infection/genetics , Zika Virus Infection/virology
7.
PLoS Negl Trop Dis ; 13(11): e0007837, 2019 11.
Article in English | MEDLINE | ID: mdl-31765380

ABSTRACT

One of the most important clinical signs of dengue virus infection is the reduction of white blood cells and platelets in human peripheral blood (leukopenia and thrombocytopenia, respectively), which may significantly impair the clearance of dengue virus by the immune system. The cause of thrombocytopenia and leukopenia during dengue infection is still unknown, but may be related to severe suppression of bone marrow populations including hematopoietic stem cells and megakaryocytes, the progenitors of white blood cells and platelets respectively. Here, we explored the possibility that bone marrow suppression, including ablation of megakaryocyte populations, is caused by dengue virus infection of megakaryocytes. We used three different models to measure dengue virus infection and replication: in vitro, in a human megakaryocyte cell line with viral receptors, ex vivo, in primary human megakaryocytes, and in vivo, in humanized mice. All three systems support dengue virus infection and replication, including virus strains from serotypes 1, 2, and 3, and clinical signs, in vivo; all assays showed viral RNA and/or infectious viruses 7-14 days post-infection. Although we saw no significant decrease in cell viability in vitro, there was significant depletion of mature megakaryocytes in vivo. We conclude that megakaryocytes can produce dengue viruses in the bone marrow niche, and a reduction of cell numbers may affect bone marrow homeostasis.


Subject(s)
Dengue Virus/growth & development , Dengue/pathology , Megakaryocytes/virology , Animals , Bone Marrow/virology , Cells, Cultured , Disease Models, Animal , Female , Humans , Male , Mice , Mice, SCID , Models, Theoretical
8.
J Am Chem Soc ; 141(17): 6832-6836, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31017399

ABSTRACT

Flaviviruses, including dengue, West Nile and recently emerged Zika virus, are important human pathogens, but there are no drugs to prevent or treat these viral infections. The highly conserved Flavivirus NS2B-NS3 protease is essential for viral replication and therefore a drug target. Compound screening followed by medicinal chemistry yielded a series of drug-like, broadly active inhibitors of Flavivirus proteases with IC50 as low as 120 nM. The inhibitor exhibited significant antiviral activities in cells (EC68: 300-600 nM) and in a mouse model of Zika virus infection. X-ray studies reveal that the inhibitors bind to an allosteric, mostly hydrophobic pocket of dengue NS3 and hold the protease in an open, catalytically inactive conformation. The inhibitors and their binding structures would be useful for rational drug development targeting Zika, dengue and other Flaviviruses.


Subject(s)
Antiviral Agents/therapeutic use , Protease Inhibitors/therapeutic use , Serine Endopeptidases/metabolism , Viral Nonstructural Proteins/antagonists & inhibitors , Zika Virus Infection/drug therapy , Allosteric Site , Aminopyridines/chemical synthesis , Aminopyridines/metabolism , Aminopyridines/therapeutic use , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Crystallography, X-Ray , Dengue Virus/enzymology , Drug Discovery , Female , Humans , Male , Mice, Inbred C57BL , Protease Inhibitors/chemical synthesis , Protease Inhibitors/metabolism , Protein Binding , Pyrazines/chemical synthesis , Pyrazines/metabolism , Pyrazines/therapeutic use , Serine Endopeptidases/chemistry , Vero Cells , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , West Nile virus/enzymology , Zika Virus/enzymology
9.
PLoS Negl Trop Dis ; 12(5): e0006439, 2018 05.
Article in English | MEDLINE | ID: mdl-29771921

ABSTRACT

Mosquito saliva is a very complex concoction of >100 proteins, many of which have unknown functions. The effects of mosquito saliva proteins injected into our skin during blood feeding have been studied mainly in mouse models of injection or biting, with many of these systems producing results that may not be relevant to human disease. Here, we describe the numerous effects that mosquito bites have on human immune cells in mice engrafted with human hematopoietic stem cells. We used flow cytometry and multiplex cytokine bead array assays, with detailed statistical analyses, to detect small but significant variations in immune cell functions after 4 mosquitoes fed on humanized mice footpads. After preliminary analyses, at different early times after biting, we focused on assessing innate immune and subsequent cellular responses at 6 hours, 24 hours and 7 days after mosquito bites. We detected both Th1 and Th2 human immune responses, and delayed effects on cytokine levels in the blood, and immune cell compositions in the skin and bone marrow, up to 7 days post-bites. These are the first measurements of this kind, with human immune responses in whole animals, bitten by living mosquitoes, versus previous studies using incomplete mouse models and salivary gland extracts or needle injected saliva. The results have major implications for the study of hematophagous insect saliva, its effects on the human immune system, with or without pathogen transmission, and the possibility of determining which of these proteins to target for vaccination, in attempts to block transmission of numerous tropical diseases.


Subject(s)
Aedes/immunology , Immune System/immunology , Insect Bites and Stings/immunology , Saliva/immunology , Aedes/physiology , Animals , Cytokines/genetics , Cytokines/immunology , Female , Humans , Immune System/parasitology , Insect Bites and Stings/genetics , Insect Bites and Stings/parasitology , Insect Proteins/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Natural Killer T-Cells/immunology
10.
J Infect Dis ; 217(4): 538-547, 2018 01 30.
Article in English | MEDLINE | ID: mdl-28968863

ABSTRACT

Background: While Zika virus (ZIKV) is mainly transmitted by mosquitoes, numerous cases of sexual transmission have been reported during recent outbreaks. Little is known about which host cell types or entry factors aid in mediating this sexual transmission. Methods: In this study, we investigated ZIKV cell tropism by infecting 2 types of human prostate cells with 3 contemporary ZIKV isolates from persons infected in the Americas. We used real-time quantitative polymerase chain reaction and immunofluorescence analyses to measure infection and flow cytometry to detect entry factor expression. Results: Here we show that ZIKV infects, replicates, and produces infectious virus in prostate stromal mesenchymal stem cells, epithelial cells, and organoids made with a combination of these cells. We also show that prostate cells express several well-characterized flavivirus attachment factors. In contrast, dengue virus does not infect or does not replicate in these prostate cells, although it is known to use similar receptors. Conclusions: Our results indicate that ZIKV favors infection of stromal cells more so than epithelial cells in organoids, possibly indicating a preference for stem cells in general. Overall, these results suggest that ZIKV replication occurs in the human prostate and can account for ZIKV secretion in semen, thus leading to sexual transmission.


Subject(s)
Epithelial Cells/virology , Mesenchymal Stem Cells/virology , Prostate/virology , Viral Tropism , Virus Replication , Zika Virus/physiology , Americas , Flow Cytometry , Humans , Male , Microscopy, Fluorescence , Real-Time Polymerase Chain Reaction , Virus Cultivation , Zika Virus/isolation & purification , Zika Virus Infection/virology
11.
J Gen Virol ; 98(8): 2106-2112, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28786787

ABSTRACT

We propose the creation of seven new species in the genus Pestivirus (family Flaviviridae) in addition to the four existing species, and naming species in a host-independent manner using the format Pestivirus X. Only the virus species names would change; virus isolates would still be referred to by their original names. The original species would be re-designated as Pestivirus A (original designation Bovine viral diarrhea virus 1), Pestivirus B (Bovine viral diarrhea virus 2), Pestivirus C (Classical swine fever virus) and Pestivirus D (Border disease virus). The seven new species (and example isolates) would be Pestivirus E (pronghorn pestivirus), Pestivirus F (Bungowannah virus), Pestivirus G (giraffe pestivirus), Pestivirus H (Hobi-like pestivirus), Pestivirus I (Aydin-like pestivirus), Pestivirus J (rat pestivirus) and Pestivirus K (atypical porcine pestivirus). A bat-derived virus and pestiviruses identified from sheep and goat (Tunisian sheep pestiviruses), which lack complete coding region sequences, may represent two additional species.


Subject(s)
Pestivirus Infections/veterinary , Pestivirus/classification , Pestivirus/isolation & purification , Animals , Cattle , Goats , Pestivirus/genetics , Pestivirus/physiology , Pestivirus Infections/virology , Phylogeny , Rats , Sheep , Swine
12.
J Gen Virol ; 98(1): 2-3, 2017 01.
Article in English | MEDLINE | ID: mdl-28218572

ABSTRACT

The Flaviviridae is a family of small enveloped viruses with RNA genomes of 9000-13 000 bases. Most infect mammals and birds. Many flaviviruses are host-specific and pathogenic, such as hepatitis C virus in the genus Hepacivirus. The majority of known members in the genus Flavivirus are arthropod borne, and many are important human and veterinary pathogens (e.g. yellow fever virus, dengue virus). This is a summary of the current International Committee on Taxonomy of Viruses (ICTV) report on the taxonomy of the Flaviviridae, which is available at www.ictv.global/report/flaviviridae.


Subject(s)
Flaviviridae/classification , Animals , Arthropod Vectors/virology , Flaviviridae/genetics , Flaviviridae/physiology , Flaviviridae/ultrastructure , Flaviviridae Infections/transmission , Flaviviridae Infections/veterinary , Flaviviridae Infections/virology , Humans
13.
Sci Rep ; 7: 41389, 2017 01 27.
Article in English | MEDLINE | ID: mdl-28128342

ABSTRACT

Zika virus (ZIKV) is an emerging mosquito-borne (Aedes genus) arbovirus of the Flaviviridae family. Although ZIKV has been predominately associated with a mild or asymptomatic dengue-like disease, its appearance in the Americas has been accompanied by a multi-fold increase in reported incidence of fetal microcephaly and brain malformations. The source and mode of vertical transmission from mother to fetus is presumptively transplacental, although a causal link explaining the interval delay between maternal symptoms and observed fetal malformations following infection has been missing. In this study, we show that primary human placental trophoblasts from non-exposed donors (n = 20) can be infected by primary passage ZIKV-FLR isolate, and uniquely allowed for ZIKV viral RNA replication when compared to dengue virus (DENV). Consistent with their being permissive for ZIKV infection, primary trophoblasts expressed multiple putative ZIKV cell entry receptors, and cellular function and differentiation were preserved. These findings suggest that ZIKV-FLR strain can replicate in human placental trophoblasts without host cell destruction, thereby serving as a likely permissive reservoir and portal of fetal transmission with risk of latent microcephaly and malformations.


Subject(s)
Placenta/pathology , Trophoblasts/virology , Virus Replication/physiology , Zika Virus/physiology , Adult , Cells, Cultured , Dengue/pathology , Dengue/virology , Dengue Virus/physiology , Female , Giant Cells/metabolism , Giant Cells/pathology , Humans , Ligands , MicroRNAs/genetics , MicroRNAs/metabolism , Phylogeny , Pregnancy , RNA, Viral/metabolism , Receptors, Virus/metabolism , Toll-Like Receptors/metabolism , Zika Virus Infection/pathology , Zika Virus Infection/virology
15.
J Gen Virol ; 97(11): 2894-2907, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27692039

ABSTRACT

Proposals are described for the assignment of recently reported viruses, infecting rodents, bats and other mammalian species, to new species within the Hepacivirus and Pegivirus genera (family Flaviviridae). Assignments into 14 Hepacivirus species (Hepacivirus A-N) and 11 Pegivirus species (Pegivirus A-K) are based on phylogenetic relationships and sequence distances between conserved regions extracted from complete coding sequences for members of each proposed taxon. We propose that the species Hepatitis C virus is renamed Hepacivirus C in order to acknowledge its unique historical position and so as to minimize confusion. Despite the newly documented genetic diversity of hepaciviruses and pegiviruses, members of these genera remain phylogenetically distinct, and differ in hepatotropism and the possession of a basic core protein; pegiviruses in general lack these features. However, other characteristics that were originally used to support their division into separate genera are no longer definitive; there is overlap between the two genera in the type of internal ribosomal entry site and the presence of miR-122 sites in the 5' UTR, the predicted number of N-linked glycosylation sites in the envelope E1 and E2 proteins, the presence of poly U tracts in the 3' UTR and the propensity of viruses to establish a persistent infection. While all classified hepaciviruses and pegiviruses have mammalian hosts, the recent description of a hepaci-/pegi-like virus from a shark and the likely existence of further homologues in other non-mammalian species indicate that further species or genera remain to be defined in the future.


Subject(s)
Flaviviridae Infections/veterinary , Flaviviridae Infections/virology , Flaviviridae/classification , Hepacivirus/classification , Hepatitis C/veterinary , Hepatitis C/virology , Animals , Chiroptera/virology , Flaviviridae/genetics , Flaviviridae/isolation & purification , Genetic Variation , Hepacivirus/genetics , Hepacivirus/isolation & purification , Humans , Mammals/virology , Phylogeny , Rodentia/virology , Sequence Analysis, DNA
16.
PLoS Negl Trop Dis ; 10(9): e0005019, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27654889

ABSTRACT

BACKGROUND: Zika virus (Flavivirus genus) is the first mosquito-borne virus known to cause high rates of microcephaly and abortion in humans. Typically, Zika virus causes a self-limiting, systemic illness; however, the current outbreak of Zika virus in the Americas has been associated with increased rates of fetal malformations and Guillain-Barré syndrome. Very few Zika virus isolates have been described in the literature, and live viruses are needed to perform studies of pathogenesis and to develop vaccines and treatments. METHODOLOGY/CLINICAL FINDINGS: We isolated Zika virus, strain FLR, directly from the serum of an individual infected in Barranquilla, Colombia (December, 2015). Here, we describe the patient's clinical course and characterize strain FLR by its growth characteristics in mosquito and mammalian cells and its partial resistance to UV-inactivation. The full genome sequence of FLR was also analyzed (including the 3' un-translated region), to determine its probable geographic origin, and to pinpoint structural differences from other Zika virus strains. CONCLUSIONS/SIGNIFICANCE: We anticipate that the study of this low passage, clinical isolate of Zika virus, which is available for worldwide distribution, will help uncover the mechanisms of viral replication and host immune responses contributing to the varied and sometimes severe clinical presentations seen during the current epidemic in the Americas.

SELECTION OF CITATIONS
SEARCH DETAIL
...