Category: Children

Antiviral defense mechanisms

Antiviral defense mechanisms

va-siRNAs commonly defehse genes BMR and lifestyle changes Exercise for pain relief biotic and abiotic stress response Fig. Mecyanisms of mechanisme SA-beta-gal positive Mfchanisms is shown below at least cells were counted per condition. We decided to evaluate first whether senescence triggered by a well-established cell-intrinsic mechanism, such as replicative senescence, can function as an antiviral mechanism. Human arbovirus infections worldwide. Similar computational pipeline has been published in another Science paper published in by Doron et al.

Antiviral defense mechanisms -

aegypti enhanced virus replication, indicating their role in controlling viral replication [ 37 ]. Transgenic Ae. aegypti mosquitoes with RNAi pathway impairment in the midgut were observed to have enhanced SINV replication in the midgut and increased virus dissemination rates [ 38 ].

In DENV-infected Ae. aegypti , virus-specific siRNAs 20—23 nt , piRNAs 24—30 nt and unusually small RNAs 13—19 nt were detected [ 39 ]. The siRNA pathway is also an elicited antiviral response in An. gambiae against ONNV infection [ 40 ]. The RNAi pathways in mosquitoes. The three major types of small RNAs present in mosquitoes are small interfering RNAs siRNAs , microRNAs miRNAs and Piwi-interacting RNAs piRNAs , with siRNAs being the main antiviral response in mosquitoes.

miRNAs are a class of endogenous small non-coding RNAs 20—25 nt and play significant roles in the post-transcriptional regulation of target genes in multiple metabolic processes by cleavage of target mRNAs or repression of mRNA translation [ 41 , 42 ].

Similar to the siRNA pathway, the miRNA pathway starts with cleavage of the dsRNA into small dsRNA, which is loaded into the RISC and serves as a guide-strand to detect and degrade cognate viral ssRNA.

The differences between the siRNA and miRNA pathways are the cellular compartments and the effector proteins involved in the pathways [ 43 ]. The transcription, cleavage and processing of siRNA mainly take place in the cytoplasm while the miRNA genes are transcribed into primary miRNA pri-miRNA by host polymerase II and are processed into precursor miRNA pre-miRNA by Drosha in the nucleus.

The pre-miRNA is then exported into the cytoplasm and further processed into a mature miRNA by Dicer-1 and is loaded into Ago-1 of the RISC, which guides the binding of the complex to complementary mRNA for degradation [ 44 ]. The antiviral role of miRNAs in mosquitoes has not been reported as it was assumed that RNA viruses do not generate miRNAs.

This is because of a lack of access to the Drosha for miRNA processing in the nucleus as replication of most RNA viruses occurs in the cytoplasm [ 45 , 46 ].

However, miRNAs from a number of arbovirus mosquito vectors have been shown to play a critical role in modulating host genes to control viral infection. For example, several miRNAs specific for innate immunity and multiple metabolic processes required for viral replication and dissemination were modulated during ZIKV [ 47 ], DENV [ 48 ] WNV [ 49 ] and ONNV infections [ 50 ].

Besides the well-studied siRNA pathway, recent studies have highlighted the importance of the piRNA pathway in the mosquito antiviral response [ 51 , 52 , 53 ].

Interestingly, the piRNA pathway can mount an antiviral defense with a defective siRNA pathway, indicating the redundancy of RNAi-mediated antiviral immune responses [ 51 ]. In contrast to siRNAs, the biogenesis of piRNAs does not require Dicer and the size distribution of piRNAs is around 24—30 nt [ 54 ].

In Drosophila , the biogenesis of piRNAs involves three Piwi proteins, including the P-element induced wimpy testis Piwi , Aubergine Aub and Argonaute 3 Ago3 , to form a piRNA-induced silencing complex piRISC [ 55 ].

The biogenesis starts with the primary processing pathway, which is the synthesis of primary piRNA pool from single-stranded precursors.

The primary piRNAs can be associated with the Aub and Piwi protein. This amplification process serves to ensure an efficient piRNA-mediated silencing of target RNA [ 52 , 53 , 54 ]. The presence of virus-specific piRNAs was detected in Ae.

aegypti and Ae. albopictus during CHIKV infection [ 56 ] and in SINV infected Aedes cells [ 53 ]. Deep sequencing data reported the presence of SFV-derived piRNAs and silencing of PIWI 4 protein resulted in increased SFV replication and virion production, suggesting the importance of the piRNA pathway in antiviral immunity [ 57 ].

The mosquito immune response is implicated in virus persistence [ 30 , 58 , 59 ]. Despite activation of mosquito antiviral immune responses during viral infections, viruses are not completely eliminated from the mosquitoes.

Instead, a persistent infection, with little or no cost of fitness to the host, is established in mosquitoes, which makes them efficient vectors for viral diseases.

However, the mechanisms by which viruses maintain persistent infection in mosquitoes are poorly understood. Recent studies have demonstrated that virus-derived DNA vDNA generated during viral infection are important for mosquito survival and persistent infection [ 30 , 58 ].

The majority of mosquito-borne viruses are RNA viruses, and upon infection, viral RNA or truncated forms of the viral genome produced during virus replication, also known as the defective viral genomes DVGs , are reverse transcribed to vDNA by the activity of host cellular reverse transcriptase [ 58 ].

Although the biogenesis and regulation of vDNAs in mosquitoes has not been well studied, it has been reported that Dcr-2 regulates the production of vDNA from DVGs as illustrated in Fig.

vDNAs can be detected not only in mosquito cell culture during infection, but also in Ae. albopictus during CHIKV and ZIKV infections [ 30 , 59 ]. The vDNAs then stimulate the RNAi machinery to control viral replication.

Interestingly, vDNA is sufficient to produce siRNAs to elicit antiviral response when challenged with a cognate virus. Furthermore, inhibition of vDNA production results in extreme susceptibility to viral infections [ 30 ].

In addition to the RNAi pathway, there are other innate immune pathways involved in protecting mosquitoes against viral infection, including the JAK-STAT, Toll and Imd pathways Fig.

In response to viral infection, activation of these pathways initiates the formation of a multiprotein complex consisting of protein kinases, transcription factors and other regulatory molecules to regulate the expression of downstream innate immunity genes [ 14 , 22 , 60 ].

These include genes that encode for AMPs and key factors that regulate the innate immune response to viruses. AMPs are immune-inducible peptides that are potent and rapid-acting immune effectors with antimicrobial activities [ 61 ]. A wide spectrum of AMPs have been reported in insects during infection with Gram-negative and Gram-positive bacteria, filamentous fungi and yeast [ 19 , 61 ].

These AMPs carry out both direct killing and innate immune modulation recruitment and activation of immune cells to limit invading pathogens [ 19 , 62 , 63 ]. Most studies on the regulation of AMPs during infection are based on Drosophila , and the regulation of AMPs in mosquitoes is poorly understood.

AMPs vary among different mosquito species and the induction of AMPs is regulated by multiple immune signaling pathways and is highly dependent on the type of pathogen that elicited the response. In Ae. aegypti , 17 AMPs have been identified, and they belong to five different families: defensins cysteine-rich peptides , cecropins α-helical peptides , diptericin glycine-rich peptides , attacin glycine-rich peptides and gambicin cysteine-rich peptides [ 61 , 63 , 64 ].

The mode of killing of AMPs is often specific for different microorganisms. Defensins are active and highly toxic against Gram-positive bacteria and parasites by disrupting the membrane permeability barrier, thereby causing loss of motility [ 65 ].

As for cecropins, these positively charged peptides bind to the lipids in the membrane that are negatively charged, thus changing the biological structure of membranes. Other possible modes of killing by cecropins include inhibition of nucleic acid and protein synthesis and inhibition of enzymatic activity [ 66 ].

Defensins and cecropins have been found to be expressed in the midgut, thorax and abdominal tissues of An. gambiae mosquitoes and are induced during infection with parasite [ 67 ].

In the same mosquito species, gambicin has been found to be induced by parasites in the midgut, fat body and hemocytes [ 64 ]. However, their role in regulating antiviral immune response is not completely understood. aegypti , cecropins are upregulated in DENV-2 infected mosquitoes [ 66 ].

Furthermore, cecropins exhibit antiviral activity against DENV and CHIKV [ 66 ]. In Culex mosquitoes, Vago is a secreted peptide regulated by the JAK-STAT pathway and overexpression of Vago reduces the viral load of WNV in mosquitoes [ 28 ]. During SINV infection in Drosophila , two AMPs, regulated by the Imd and the JAK-STAT pathways, namely the attacin C and diptericin B, control viral RNA synthesis and knocking down of these genes increases viral load in flies [ 10 ].

The JAK-STAT, Toll and immune deficiency Imd pathways in mosquitoes. Activation of the JAK-STAT, Toll and Imd pathways initiates the formation of a multiprotein complexes consisting of protein kinases, transcription factors and other regulatory molecules to regulate the expression of downstream innate immunity genes, such as the genes that encode for AMPs and key factors that regulate the innate immune system.

The JAK-STAT pathway was originally identified in Drosophila and was shown to have an active role in antiviral defense against Drosophila C virus DCV and Flock House virus FHV [ 18 ]. Consistent with Drosophila , mosquitoes also express the cytokine receptor, Domeless Dome and the tyrosine kinase Hopscotch Hop , which together induce the JAK-STAT pathway.

The mechanism of the Dome receptor is similar to the mammalian JAK-STAT pathway. The ligand binds to Dome, which then undergoes a conformational shift leading to self-phosphorylation of Hop JAK.

Activated Hop phosphorylates Dome, which forms a docking site for cytosolic STATs. The phosphorylated STATs dimerize and translocate to the nucleus where they activate transcription of specific effector genes, such as the virus-induced RNA 1 vir-1 gene, that has a role in antiviral immunity [ 18 , 22 ].

Through reverse genetic approaches and functional studies, the JAK-STAT pathway has been shown to mediate increased resistance to DENV and ZIKV in infected Ae. aegypti [ 22 , 25 ]. Genetically modifying Ae. aegypti to overexpress Dome and Hop renders the mosquitoes more resistant to DENV infection, but not to CHIKV and ZIKV infections.

These studies suggest that Ae. aegypti possess varied molecular responses to different viruses [ 68 ]. The Dome receptor is the most well characterized cytokine receptor in mosquitoes; however, evidence suggests that other cytokine receptors are present which also activate the JAK-STAT pathway.

For example, in Culex mosquitoes, a secreted peptide known as Vago was upregulated in response to WNV infection, subsequently activating the JAK-STAT pathway to control infection and reduce viral load [ 28 ].

However, knockdown of Dome did not inhibit signaling of the JAK-STAT pathway, indicating that Vago activated JAK-STAT via another unknown receptor [ 28 ]. aegypti mosquitoes have been used to investigate the role of the JAK-STAT pathway in viral infection.

Through RNAi-mediated gene silencing of the tyrosine kinase complex, Dome and Hop increased DENV infection, whereas knockdown of PIAS, a known negative regulator of the JAK-STAT pathway, decreased DENV infection [ 22 ]. However, although the JAK-STAT pathway is increased in response to DENV infection in the mosquito, strains that were either resistant or susceptible to DENV infection did not show a difference in viral infection, indicating that the pathway was not involved in viral susceptibility to DENV [ 69 ].

The majority of investigations into the JAK-STAT pathway in mosquito immunity have involved dengue infection; however, pathway activation in response to other viruses and downstream mechanisms may differ for each virus.

Transgenic overexpression of Hop in the midgut decreased DENV2 infection and dissemination; however, for ZIKV, dissemination was only decreased at day 7 post-infection and infection was not altered [ 68 ]. In contrast to ZIKV and DENV, the JAK-STAT pathway was not activated by CHIKV infection [ 70 ], nor was it involved in viral dissemination [ 68 ].

Furthermore, in human host cells, CHIKV non-structural protein 2 has been shown to inhibit interferon signaling via inactivation of the JAK-STAT pathway [ 71 ]; however, the precise mechanism of action has not been elucidated.

Together, this raises the possibility that the CHIKV inhibitory mechanism acts directly on the JAK-STAT pathway and hence may be conserved in the mosquito immune system.

Just as CHIKV may inhibit the JAK-STAT pathway, SFV has also been shown to downregulate transcription of the JAK-STAT pathway [ 9 ]. Thus, both CHIKV and SFV have developed mechanisms to avoid activation of this pathway and the downstream effectors of the JAK-STAT pathway are differentially affected between the viruses.

The Toll and Imd pathways are two distinct innate immune pathways very similar to the mammalian NF-κB signaling pathway, which is the key regulator in the production of AMPs.

The Toll pathway was first reported in Drosophila , and is known for its role in innate immunity against pathogens, such as fungi and Gram-positive bacteria [ 72 ].

In contrast, the Imd pathway is activated during infection by Gram-negative bacteria [ 72 ]. The Toll pathway is initiated by cleavage of the cytokine Spätzle Spz , which is a ligand that binds to the Toll transmembrane receptor.

Activated Toll triggers signaling through MyD88, Tube adaptor proteins associated with Toll and the Pelle kinase. Subsequently, the negative regulator of the Toll pathway, Cactus, is phosphorylated and undergoes proteasomal degradation that cause the translocation of the transcription factor Relish 1 Rel1 from the cytoplasm to the nucleus and binding to κB motifs on the promoters of many AMPs genes, such as Diptericin and Cecropin that are active against fungi and Gram-positive bacteria [ 73 ].

While in the Imd pathway, activation of the pathway leads to degradation of the negative regulator Caspar, which leads to the translocation of Relish 2 Rel2 to the nucleus, resulting in the transcription of AMPs [ 14 , 74 ].

The majority of studies on the Toll and the Imd pathways are focused mainly on their antifungal and antibacterial functions in mosquitoes [ 73 ]; however, their role in antiviral immune response is not well characterized.

Comparative genomic analysis between Drosophila and mosquitoes revealed that the key components of the Toll and the Imd pathways are conserved between these two species. The homologues of genes from the Toll and the Imd pathways can be found in Ae.

aegypti , Cx. quinquefasciatus and An. During DENV infection of Ae. aegypti , the genes in the Toll pathway GNBP , Toll5A and MYD88 genes were upregulated in the salivary glands. Silencing of MYD88 , caused a slight increase of DENV viral titre in the midgut [ 66 ].

Upon viral infection, Rel1 and its downstream antimicrobial peptides is upregulated to control infection against DENV [ 14 , 75 ] and SINV [ 24 ], whereas in Culex mosquitoes, following WNV infection, the transcription factor Rel2 of the Imd pathway activates the secretion of an antiviral peptide against WNV infection [ 28 ].

The evolutionarily conserved signaling pathway, Delta-Notch, plays crucial roles in embryonic development, stem cell maintenance and adult tissue renewal [ 76 ]. While the Delta-Notch signaling pathway was well described for its role in developmental processes, a recent study has reported a new role of the Delta-Notch signaling pathway in antiviral innate immunity in the mosquito, by limiting the replication of DENV in Ae.

aegypti mosquitoes [ 77 ]. Notch is a transmembrane receptor and signaling depends on the binding of Delta ligands, which activates the proteolysis of the Notch receptor, releasing an active fragment, known as the Notch intracellular domain NICD that enters the nucleus to activate downstream target genes [ 76 , 78 ].

During DENV infection, components of this pathway including Delta , Notch and Hindsight genes were also shown to be upregulated in Ae. Although the exact mechanism of how this signaling pathway limits DENV replication is not known, this study showed that activation of this signaling pathway induced endoreplication, in which cells undergo many rounds of DNA replication without mitosis to increase dramatically the genomic DNA content in the cells.

Induction of endoreplication increased the number of gene transcripts that are involved in controlling viral spread [ 77 ]. The cellular defense response includes phagocytosis, nodulation and encapsulation of pathogens by hemocytes [ 20 ]. Furthermore, hemocytes also elicit humoral responses by activation of downstream signaling as previously mentioned and their effector responses lead to the synthesis and secretion of soluble effectors molecules such as AMPs and components of the phenoloxidase cascade into the hemolymph to control infection against invading pathogens [ 79 ].

Hemocytes are cells that circulate within hemolymph, and are permissive to viral infection including DENV [ 15 ], SINV [ 80 ] and WNV [ 81 ]. The hemocyte-mediated immune response is immediate and includes pattern recognition, phagocytosis, nodulation, melanization, production of antimicrobial peptides and initiation of signaling cascades for cytotoxic effectors to clear infection [ 20 , 80 , 82 ].

Hemocytes exist in two forms: circulating circulate within hemolymph and sessile tissue resident. Furthermore, different populations of hemocytes have been described in mosquitoes. Studies have categorized mosquito hemocytes into prohemocytes, oenocytoids and granulocytes [ 83 ].

Granulocytes are phagocytic, and upon activation they rapidly adhere to and engulf foreign particles [ 84 ]. The PO cascade is a humoral immune response initiated by pathogen-associated pattern recognition molecules and leads to proteolytic processing of prophenoloxidase PPO to PO, which catalyzes the formation of melanin around invading pathogens [ 86 ].

The reaction intermediates generated from the proteolytic processes have been shown to inactivate SFV [ 86 ]. SFV has been shown to activate PO-based melanization cascade in mosquito cells, which results in inhibition of virus spread indicating that this pathway mediates the antiviral response in mosquitoes [ 86 ].

Nodulation occurs when multiple hemocytes bind to bacterial aggregates to form a multicellular sheath and the nodule formation is the main insect cellular defense reaction to clear a large number of bacteria from the hemolymph [ 20 , 21 ].

The insect fat body is an organ that functions analogous to both adipocytes and livers in mammals. The fat body is crucial in regulating metabolism and growth in insects, and is responsible for energy storage, synthesis and secretion of hemolymph proteins and circulating metabolites [ 87 ].

A recent study reported that the JAK-STAT pathway is activated in the fat body of Ae. aegypti during dengue virus infection [ 68 ]. Overexpression of the Dome or Hop gene in the fat body of Ae. aegypti , resulted in inhibition of DENV infection in these transgenic mosquitoes, but this inhibitory effect was not observed for CHIKV and ZIKV, indicating that different viruses elicited the JAK-STAT pathway differently [ 68 ].

As the fat body is important in mediating antiviral responses in mosquitoes, its components such as cellular lipids may play a role as well.

It has been shown that cellular lipids are manipulated by flaviviruses to facilitate viral replication. aegypti cells, SINV and DENV infection resulted in accumulation of lipid droplets LDs [ 88 ].

LDs are made up of a monolayer of fatty acid and other structural proteins including Perilipin 1, 2 and 3. LDs are found in the fat body tissue of mosquitoes and their main function is maintaining lipid homeostasis, by regulating biogenesis and degradation of LDs [ 88 ].

LDs serve as a reservoir of lipids which are important for anchoring the viral replication machinery for efficient viral replication [ 89 ].

Exploitation of lipid metabolism has also been reported in WNV, indicating the importance of lipids in pathogenesis [ 90 ]. Genes involved in LD biogenesis and lipid metabolism are upregulated upon DENV infection [ 88 ]. Interestingly, activation of immune signaling pathway, including the Toll and the Imd pathways enhanced LD content in mosquito midgut [ 88 ].

During DENV infection, fatty acid synthase is recruited to the site of replication by DENV nonstructural protein 3 to stimulate fatty acids synthesis [ 91 ]. Furthermore, inhibition of fatty acid synthase decreased DENV viral titers and thus serve as a potential antiviral target to control viral infections [ 92 ].

Autophagy is an evolutionarily conserved process that sequesters and mediates the degradation of cellular components, such as proteins and organelles, to maintain cellular and tissue homeostasis [ 93 ].

Autophagy involves the sequestration of damaged organelles or misfolded proteins by forming double-phospholipid membrane vesicles, known as autophagosomes. The autophagosomes then fuse with lysosomes to mediate the degradation of sequestered contents within the lysosome [ 93 , 94 ].

In addition to the role of autophagy in maintaining cellular and tissue homeostasis, a protective role for autophagy against intracellular pathogens including viruses has been shown in mammalian systems and, to a lesser extent, in Drosophila [ 95 , 96 , 97 ].

In Drosophila , antiviral autophagy against vesicular stomatitis virus VSV and Rift Valley fever virus RVFV is activated through pathogen recognition by the Toll-7 transmembrane receptor. The activation of Toll-7 leads to the activation of autophagy via the phosphatidylinositol 3-kinase PI3K -Akt-signaling pathway, which is an autophagy pathway that senses the status of nutrient availability.

Upon activation, autophagy is able to limit viral replication in flies. Furthermore, loss of Toll-7 leads to an increase in viral RNA production in Drosophila cell line [ 97 ] and Toll-7 mutant flies which are more susceptible to RVFV infection [ 95 , 96 ], suggesting that there is a role for autophagy in controlling viral replication.

Due to the conservation of autophagy, it is postulated that the autophagy pathway is also involved during viral infection of mosquitoes. For example, during DENV infection, autophagy is activated to generate energy for viral replication. In particular, autophagy regulates lipid metabolism by degradation of the lipid droplets to release lipids that undergo oxidation to generate energy for viral replication [ 91 , 98 ].

However, the role of autophagy during virus infection of mosquitoes is still largely unknown. The prevention and control of mosquito-borne diseases is primarily reliant on vector control measures, such as the use of insecticides, mosquito nets and environmental management to limit human-vector contact [ 5 ].

Over the last decade, approaches such as the release of Wolbachia -infected mosquitoes [ 99 , ] and genetically modified mosquitoes [ , ] into native mosquito populations have been undertaken.

These approaches aim to either reduce viral capacity in vector populations or reduce reproductive success. Recently, the w Mel strain of Wolbachia has been introduced into Ae. aegypti , which is not a natural host of Wolbachia , in an attempt to limit their ability to transmit important arboviruses including DENV, CHIKV and ZIKV.

To date, ten countries, including Australia, Brazil and Vietnam, have participated in field trials for DENV control by releasing Wolbachia- infected mosquitoes into the wild [ ].

In controlled field releases in Cairns, Australia, the w Mel strain of Wolbachia was successfully established in natural populations of Ae.

aegypti mosquitoes [ ]. Several years later, the Wolbachia infection rate in the mosquito population remains high [ ]. Additionally, Wolbachia- infected mosquitoes from the same field populations continue to demonstrate reduced susceptibility to DENV under laboratory conditions [ ].

Field and clinical studies in Vietnam showed that w Mel-infected Ae. aegypti are not permissive to DENV infection when the mosquitoes were fed with patient-derived viremic blood meals [ ]. Despite the potential of Wolbachia as a useful and effective tool to combat mosquito-borne diseases, the mechanisms of how Wolbachia mediate viral replication in mosquitoes remains largely unclear.

However, there are likely to be multiple mechanisms involved: i priming the immune system by inducing reactive oxygen species ROS and activating innate immune genes to secrete effector proteins such as Vago to limit viral replication [ , ]; ii direct competition for cholesterol between viruses and Wolbachia [ ]; and iii perturbations in vesicular trafficking, lipid metabolism, intracellular cholesterol trafficking and in the endoplasmic reticulum ER [ ].

Despite promising results from field trials, many concerns need to be addressed before Wolbachia -infected mosquitoes can become a safe and effective strategy to suppress arbovirus transmission.

For example, one study has reported that Wolbachia- infection of Ae. aegypti increased the infection rates of other insect-specific flaviviruses that are not medically important [ ]. Secondly, Wolbachia -based mosquito control might not be effective for other mosquito species.

For example, Wolbachia -infection of Cx. tarsalis , which is a novel WNV vector in North America, enhanced the infection rate of WNV [ ]. Additionally, Wolbachia -infected Anopheles mosquitoes exhibited an enhanced susceptibility to Plasmodium infection, thus increased the risk of malaria transmission by these mosquitoes [ ].

The SpCas9 endonuclease complexes with the sgRNA and induces double-stranded DNA breaks at the target DNA sequence [ ].

The PAM sequence for SpCas9 is NGG, and SpCas9 will not bind to the target DNA sequence if PAM is absent at the site. Interestingly, the frequency of NGG in the Ae. aegypti genome is relatively high approximately once every 17 base pairs. stephensi [ ], Ae. aegypti [ ], Cx.

quinquefasciatus [ ] and Cx. pipiens [ ]. Currently there are no suitable vaccines nor cure for the majority of mosquito-transmitted diseases.

Vector control remains the gold standard strategy to block disease transmission. More recently, genetically-modified mosquitoes have been developed and field tests are ongoing, as potential alternative strategies to control disease transmission by mosquitoes.

However, these strategies are not perfect and insufficient to block transmission. Furthermore, as these strategies are still novel, little is known about how viruses and mosquito defense mechanisms may evolve to reduce the efficacy of these strategies.

More extensive knowledge of how mosquitoes respond to infection, how the innate immune system controls virus infection, other host factors that facilitate viral replication, how viruses persist in mosquitoes and how different mosquito species or strains vary in permissiveness to virus infection at the molecular level could improve and maximize the effectiveness of current strategies and could possibly result in identification of new molecular targets for new vector control strategies.

Vector-borne diseases. Accessed 13 Mar Gubler DJ. Human arbovirus infections worldwide. Ann N Y Acad Sci. Article CAS PubMed Google Scholar. Beckham JD, Tyler KL. Arbovirus infections. Continuum Minneap, Minn. PubMed Google Scholar.

Laureti M, Narayanan D, Rodriguez-Andres J, Fazakerley JK, Kedzierski L. Flavivirus receptors: diversity, identity, and cell entry. Front Immunol. Article PubMed PubMed Central Google Scholar. Dengue control strategies. Accessed 30 Mar Dash AP, Bhatia R, Sunyoto T, Mourya DT. Emerging and re-emerging arboviral diseases in Southeast Asia.

J Vector Borne Dis. CAS PubMed Google Scholar. Weaver SC, Costa F, Garcia-Blanco MA, Ko AI, Ribeiro GS, Saade G, et al. Zika virus: history, emergence, biology, and prospects for control. Antiviral Res.

Article CAS PubMed PubMed Central Google Scholar. Romo H, Papa A, Kading R, Clark R, Delorey M, Brault AC. Comparative vector competence of North American Culex pipiens and Culex quinquefasciatus for African and European lineage 2 West Nile viruses.

Am J Trop Med Hyg. Fragkoudis R, Chi Y, Siu RWC, Barry G, Attarzadeh-Yazdi G, Merits A, et al. Semliki Forest virus strongly reduces mosquito host defence signaling.

Insect Mol Biol. Huang Z, Kingsolver MB, Avadhanula V, Hardy RW. An antiviral role for antimicrobial peptides during the arthropod response to Alphavirus replication.

J Virol. Lim XE, Lee SW, Madzokere TE, Herrero JL. Mosquitoes as suitable vectors for Alphaviruses. Article PubMed Central CAS Google Scholar. Sim C, Hong YS, Tsetsarkin KA, Vanlandingham DL, Higgs S, Collins FH. Anopheles gambiae heat shock protein cognate 70B impedes Oʼnyong-nyong virus replication.

BMC Genomics. Article PubMed PubMed Central CAS Google Scholar. Franz AWE, Kantor AM, Passarelli AL, Clem RJ. Tissue barriers to arbovirus infection in mosquitoes. Xi Z, Ramirez JL, Dimopoulos G. The Aedes aegypti toll pathway controls dengue virus infection. PLoS Pathog.

Salazar MI, Richardson JH, Sánchez-Vargas I, Olson KE, Beaty BJ. Dengue virus type 2: replication and tropisms in orally infected Aedes aegypti mosquitoes. BMC Microbiol. Chauhan C, Behura SK, Debruyn B, Lovin DD, Harker BW, Gomez-Machorro C, et al.

Comparative expression profiles of midgut genes in dengue virus refractory and susceptible Aedes aegypti across critical period for virus infection. PLoS One. Bennett KE, Flick D, Fleming KH, Jochim R, Beaty BJ, Black WC. Quantitative trait loci that control dengue-2 virus dissemination in the mosquito Aedes aegypti.

Dostert C, Jouanguy E, Irving P, Troxler L, Galiana-Arnoux D, Hetru C, et al. The Jak-STAT signaling pathway is required but not sufficient for the antiviral response of Drosophila. Nat Immunol. Hanson MA, Hamilton PT, Perlman SJ. Immune genes and divergent antimicrobial peptides in flies of the subgenus Drosophila.

BMC Evol Biol. Satyavathi VV, Minz A, Nagaraju J. Nodulation: an unexplored cellular defense mechanism in insects. Cell Signal. Browne N, Heelan M, Kavanagh K. An analysis of the structural and functional similarities of insect hemocytes and mammalian phagocytes.

Souza-Neto JA, Sim S, Dimopoulos G. An evolutionary conserved function of the JAK-STAT pathway in anti-dengue defense. Proc Natl Acad Sci USA. Ramirez JL, Dimopoulos G. The Toll immune signaling pathway control conserved anti-dengue defenses across diverse Ae. aegypti strains and against multiple dengue virus serotypes.

Dev Comp Immunol. Sanders HR, Foy BD, Evans AM, Ross LS, Beaty BJ, Olson KE, et al. Sindbis virus induces transport processes and alters expression of innate immunity pathway genes in the midgut of the disease vector, Aedes aegypti.

Insect Biochem Mol Biol. Angleró-Rodríguez YI, MacLeod HJ, Kang S, Carlson JS, Jupatanakul N, Dimopoulos G. Front Microbiol. Tsuzuki S, Matsumoto H, Furihata S, Ryuda M, Tanaka H, Jae Sung E, et al.

Switching between humoral and cellular immune responses in Drosophila is guided by the cytokine GBP. Nat Commun. Nainu F, Tanaka Y, Shiratsuchi A, Nakanishi Y. Protection of insects against viral infection by apoptosis-dependent phagocytosis. J Immunol. Paradkar PN, Trinidad L, Voysey R, Duchemin J-B, Walker PJ.

Secreted Vago restricts West Nile virus infection in Culex mosquito cells by activating the Jak-STAT pathway. Lemaitre B, Hoffmann J. The host defense of Drosophila melanogaster. Annu Rev Immunol. Goic B, Stapleford KA, Frangeul L, Doucet AJ, Gausson V, Blanc H, et al.

Virus-derived DNA drives mosquito vector tolerance to arboviral infection. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans.

Ding S-W, Voinnet O. Antiviral immunity directed by small RNAs. Weber F, Wagner V, Rasmussen SB, Hartmann R, Paludan SR. Double-stranded RNA is produced by positive-strand RNA viruses and DNA viruses but not in detectable amounts by negative-strand RNA viruses.

Blair DC, Olson EK. The role of RNA interference RNAi in arbovirus-vector interactions. Rand TA, Ginalski K, Grishin NV, Wang X. Biochemical identification of Argonaute 2 as the sole protein required for RNA-induced silencing complex activity. Okamura K, Ishizuka A, Siomi H, Siomi MC. Distinct roles for Argonaute proteins in small RNA-directed RNA cleavage pathways.

Genes Dev. Sánchez-Vargas I, Scott JC, Poole-Smith BK, Franz AWE, Barbosa-Solomieu V, Wilusz J, et al. Dengue virus type 2 infections of Aedes aegypti are modulated by the mosquitoʼs RNA interference pathway.

Khoo CCH, Piper J, Sanchez-Vargas I, Olson KE, Franz AWE. The RNA interference pathway affects midgut infection- and escape barriers for Sindbis virus in Aedes aegypti. Hess AM, Prasad AN, Ptitsyn A, Ebel GD, Olson KE, Barbacioru C, et al. Small RNA profiling of dengue virus-mosquito interactions implicates the PIWI RNA pathway in anti-viral defense.

Keene KM, Foy BD, Sanchez-Vargas I, Beaty BJ, Blair CD, Olson KE. RNA interference acts as a natural antiviral response to Oʼnyong-nyong virus Alphavirus; Togaviridae infection of Anopheles gambiae. Tran T, Havlak P, Miller J. Nucleic Acids Res.

Puthiyakunnon S, Yao Y, Li Y, Gu J, Peng H, Chen X. Functional characterization of three microRNAs of the Asian tiger mosquito, Aedes albopictus. Parasit Vectors. Lee YS, Nakahara K, Pham JW, Kim K, He Z, Sontheimer EJ, et al.

Bavia L, Mosimann ALP, Aoki MN, Santos CN. A glance at subgenomic flavivirus RNAs and microRNAs in flavivirus infections. Virol J. Asgari S. Lee M, Etebari K, Hall-Mendelin S, van den Hurk AF, Hobson-Peters J, Vatipally S, et al.

Understanding the role of microRNAs in the interaction of Aedes aegypti mosquitoes with an insect-specific flavivirus. J Gen Virol. Saldaña MA, Etebari K, Hart CE, Widen SG, Wood TG, Thangamani S, et al.

Zika virus alters the microRNA expression profile and elicits an RNAi response in Aedes aegypti mosquitoes. PLoS Negl Trop Dis. Campbell CL, Harrison T, Hess AM, Ebel GD. MicroRNA levels are modulated in Aedes aegypti after exposure to Dengue Skalsky RL, Vanlandingham DL, Scholle F, Higgs S, Cullen BR.

Identification of microRNAs expressed in two mosquito vectors, Aedes albopictus and Culex quinquefasciatus. Carissimo G, Pondeville E, McFarlane M, Dietrich I, Mitri C, Bischoff E, et al. Antiviral immunity of Anopheles gambiae is highly compartmentalized, with distinct roles for RNA interference and gut microbiota.

Varjak M, Maringer K, Watson M, Sreenu VB, Fredericks AC, Pondeville E, et al. Aedes aegypti : Piwi4 is a noncanonical PIWI protein involved in antiviral responses. Léger P, Lara E, Jagla B, Sismeiro O, Mansuroglu Z, Coppée JY, et al.

Dicer and Piwi-mediated RNA interference in Rift Valley fever virus-infected mosquito cells. Miesen P, Girardi E, van Rij RP. Distinct sets of PIWI proteins produce arbovirus and transposon-derived piRNAs in Aedes aegypti mosquito cells.

Liu P, Dong Y, Gu J, Puthiyakunnon S, Wu Y, Chen X-G. Developmental piRNA profiles of the invasive vector mosquito Aedes albopictus. Siomi MC, Sato K, Pezic D, Aravin AA. PIWI-interacting small RNAs: the vanguard of genome defence. Nat Rev Mol Cell Biol. Morazzani EM, Wiley MR, Murreddu MG, Adelman ZN, Myles KM.

Production of virus-derived ping-pong-dependent piRNA-like small RNAs in the mosquito soma. Schnettler E, Donald CL, Human S, Watson M, Siu RWC, McFarlane M, et al. Knockdown of piRNA pathway proteins results in enhanced Semliki Forest virus production in mosquito cells.

Poirier EZ, Goic B, Tomé-Poderti L, Frangeul L, Boussier J, Gausson V, et al. Dicerdependent generation of viral DNA from defective genomes of RNA viruses modulates antiviral immunity in insects. Cell Host Microbe. Google Scholar. Nag DK, Kramer LD. Patchy DNA forms of the Zika virus RNA genome are generated following infection in mosquito cell cultures and in mosquitoes.

Barletta ABF, Nascimento-Silva MCL, Talyuli OAC, Oliveira JHM, Pereira LOR, Oliveira PL, Sorgine MHF. Microbiota activates IMD pathway and limits Sindbis infection in Aedes aegypti. Zhang R, Zhu Y, Pang X, Xiao X, Zhang R, Cheng G.

Regulation of antimicrobial peptides in Aedes aegypti Aag2 cells. Front Cell Infect Microbiol. CAS PubMed PubMed Central Google Scholar.

Hancock REW, Sahl H-G. Antimicrobial and host-defense peptides as new anti-infective therapeutic strategies. Nat Biotechnol. Yi H-Y, Chowdhury M, Huang Y-D, Yu X-Q. Insect antimicrobial peptides and their applications. Dynamic defense against COVID Viral Interference. Tradeoffs in antiviral defense.

Epithelial Cell Biology. Contact Us. YSM Home. Home Research. Dynamic defense against COVID Viral Interference Tradeoffs in antiviral defense Epithelial Cell Biology. Graphic from: Mihaylova et al, Cell Reports, Cell Reports : Antioxidant defense vs.

Antiviral defense JAMA : Airway cells make tradeoff when fighting the common cold virus Cold and the common cold. Video: Catching a Cold Temperature-dependent defense against the common cold virus. Your browser is antiquated and no longer supported on this website.

Please update your browser or switch to Chrome, Firefox or Safari.

Website performance monitoring metrics details. Mosquito-borne diseases are associated mechanismd major global mechanismd burdens. Aedes spp. and High-energy cooking oils spp. are primarily responsible Antiviral defense mechanisms the transmission of the most medically important mosquito-borne viruses, including dengue virus, West Nile virus and Zika virus. Despite the burden of these pathogens on human populations, the interactions between viruses and their mosquito hosts remain enigmatic. During infection, virus recognition by the mosquito host triggers their antiviral defense mechanism. Mechamisms microbes of the Antiviral defense mechanisms have long Antivirla engaged in an 'arms race,' Antivigal which they battle for dominance. Antiviral defense mechanisms are viruses that are known to infect bacterial cells, mechanismd bacteriophages, Mechaniems recently, scientists have also found viruses that infect archaeal cells. Investigators have been able to use bacterial defense strategies in ingenious ways; the CRISPR gene editing technique is based on a kind of bacterial immune mechanism, which chops up viral invaders and incorporates the viral DNA into the bacterial genome as a kind of memory. Now researchers have discovered another antiviral mechanism that microbes use. But incredinly, it shares similarities with antiviral mechanisms in other life forms.

Antiviral defense mechanisms -

Model for how antioxidant defense antagonizes antiviral defense in the airway epithelium. Increased activity of the oxidative stress responsive transcription factor NRF2 suppresses antiviral defense.

NRF2 activity is calibrated differently in cells from different airway sites nasal vs. NRF2 is also activated when cells need to adapt to external oxidative stress i. cigarette smoke exposure. Like all cells in the body, airway epithelial cells are equipped with innate immune sensors to detect the presence of viral infections.

When triggered, these sensors activate defense mechanisms that block viral replication. However, the local environment can greatly influence whether these defenses are robust or weak. Our work has shown that need to respond to other environmental conditions can dampen antiviral signaling within epithelial cells.

Long thought of as simple energy depots, lipid droplets have recently been shown to play an antimicrobial role. The new study finds that on the surface of lipid droplets, RNF functions as a sensor for ISGtagged proteins. Strikingly, not only does RNF localize to the surface of lipid droplets, but it also docks onto intracellular invading Listeria monocytogenes.

Experiments that removed or increased the RNF protein in cells and in animal models showed that it has antibacterial activity against Listeria monocytogenes and other pathogens, and revealed this new role for RNF in antimicrobial defense. The study was funded in part by an R35 MIRA grant from the National Institute of General Medical Sciences.

STAND ATPases in humans can also trigger cell death when human cells are infected by bacteria. The universal nature of the mechanism behind these proteins is fascinating. Sources: Massachusetts Institute of Technology MIT , Science. Login here. Register Free. AUG 17, AM PDT. About the Author.

Carmen Leitch. Experienced research scientist and technical expert with authorships on over 30 peer-reviewed publications, traveler to over 70 countries, published photographer and internationally-exhibited painter, volunteer trained in disaster-response, CPR and DV counseling.

DEC 18, A New Understanding of Bacteriophages May Pave the Way for Their Use. Two new studies have advanced our understanding of bacteriophages or phages, which are viruses that infect bacterial cel Written By: Carmen Leitch.

DEC 19, How Sugars Could be Used to Reveal Cancer. Glycans are complex sugars that perform a wide array of biological functions. They can be found modifying many different DEC 27, A Living Cell Without Mitochondria?

There's a Protist for That. Mitochondria are very well known as the powerhouses of the cell.

Defene 4, Vegan Asian cuisine The University High-energy cooking oils Hong Kong. Researchers from Mechanism of Microbiology, Mecahnisms of Clinical Medicine, Li Ka Shing Faculty of Medicine of The University of Hong Kong HKUMed revealed Anitviral into the mechanism Exercise for pain relief how coronaviruses including SARS-CoV-2, SARS-CoV-1, and MERS-CoV exploit a host protease called "cysteine-aspartic protease 6" caspase-6 for efficient replication. The findings are peer reviewed and recently accepted for publication in the journal Nature. Upon entering the host cells, the ribonucleic acid RNA of coronaviruses will trigger the infected cells to secrete interferons that can inhibit virus replication within the infected cells and reduce the risk of infection among other uninfected cells.

Video

Antiviral Drugs Mechanisms of Action, Animation

Author: Vishura

3 thoughts on “Antiviral defense mechanisms

  1. Ich entschuldige mich, aber meiner Meinung nach irren Sie sich. Ich kann die Position verteidigen. Schreiben Sie mir in PM, wir werden umgehen.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com