Category: Children

Antiviral defense against diseases

Antiviral defense against diseases

Is global BCG aganst trained BCAAs and immune system relevant to the progression of SARS-CoV-2 Resveratrol and inflammation Article Antivital PubMed Central CAS Diseaess Scholar Merino-Ramos T, Vázquez-Calvo Á, Casas J, Sobrino F, Saiz J-C, Martín-Acebes MA. J Virol 87 23 — Role of C-reactive protein at sites of inflammation and infection. Additionally, Wolbachia- infected mosquitoes from the same field populations continue to demonstrate reduced susceptibility to DENV under laboratory conditions [ ]. Scienceeabd J Am Acad Dermatol 15 4 Pt 1 —9.

Antiviral defense against diseases -

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.

Appl Microbiol Biotechnol. Vizioli J, Bulet P, Hoffmann JA, Kafatos FC, Müller H-M, Dimopoulos G. Gambicin: a novel immune responsive antimicrobial peptide from the malaria vector Anopheles gambiae. Bartholomay LC, Fuchs JF, Cheng LL, Beck ET, Vizioli J, Lowenberger C, et al.

Reassessing the role of defensin in the innate immune response of the mosquito, Aedes aegypti. Luplertlop N, Surasombatpattana P, Patramool S, Dumas E, Wasinpiyamongkol L, Saune L, et al. Induction of a peptide with activity against a broad spectrum of pathogens in the Aedes aegypti salivary gland, following infection with dengue virus.

Vizioli J, Bulet P, Charlet M, Lowenberger C, Blass C, Muller HM, et al. Cloning and analysis of a cecropin gene from the malaria vector mosquito, Anopheles gambiae. Jupatanakul N, Sim S, Angleró-Rodríguez YI, Souza-Neto J, Das S, Poti KE, et al.

Behura SK, Gomez-Machorro C, Harker BW, deBruyn B, Lovin DD, Hemme RR, et al. Global cross-talk of genes of the mosquito Aedes aegypti in response to dengue virus infection.

Characterization of Aedes aegypti innate-immune pathways that limit chikungunya virus replication. Fros JJ, Liu WJ, Prow NA, Geertsema C, Ligtenberg M, Vanlandingham DL, et al.

Duneau DF, Kondolf HC, Im JH, Ortiz GA, Chow C, Fox MA, et al. The Toll pathway underlies host sexual dimorphism in resistance to both Gram-negative and Gram-positive bacteria in mated Drosophila. BMC Biol. Shin SW, Bian G, Raikhel AS. A toll receptor and a cytokine, Toll5A and Spz1C, are involved in toll antifungal immune signaling in the mosquito Aedes aegypti.

J Biol Chem. Kim M, Lee JH, Lee SY, Kim E, Chung J. Caspar, a suppressor of antibacterial immunity in Drosophila. Sim S, Dimopoulos G. Dengue virus inhibits immune responses in Aedes aegypti cells. Kopan R, Ilagan MXG.

The canonical notch signaling pathway: unfolding the activation mechanism. Serrato-Salas J, Hernández-Martínez S, Martínez-Barnetche J, Condé R, Alvarado-Delgado A, Zumaya-Estrada F, et al.

De novo DNA synthesis in Aedes aegypti midgut cells as a complementary strategy to limit dengue viral replication.

Sun J, Deng W-M. Eleftherianos I, Revenis C. Role and importance of phenoloxidase in insect hemostasis. J Innate Immun. Parikh GR, Oliver JD, Bartholomay LC. A haemocyte tropism for an arbovirus.

Cheng G, Cox J, Wang P, Krishnan MN, Dai J, Qian F, et al. A C-type lectin collaborates with a CD45 phosphatase homolog to facilitate West Nile virus infection of mosquitoes. Sigle LT, Hillyer JF.

Eater and draper are involved in the periostial haemocyte immune response in the mosquito Anopheles gambiae. Castillo JC, Robertson AE, Strand MR. Characterization of hemocytes from the mosquitoes Anopheles gambiae and Aedes aegypti.

Hillyer JF, Strand MR. Mosquito hemocyte-mediated immune responses. Curr Opin Insect Sci. Nakatogawa SI, Oda Y, Kamiya M, Kamijima T, Aizawa T, Clark KD, et al. A novel peptide mediates aggregation and migration of hemocytes from an insect. Curr Biol. Rodriguez-Andres J, Rani S, Varjak M, Chase-Topping ME, Beck MH, Ferguson MC, et al.

Phenoloxidase activity acts as a mosquito innate immune response against infection with Semliki Forest virus. Martins GF, Serrão JE, Ramalho-Ortigão JM, Pimenta PFP. A comparative study of fat body morphology in five mosquito species. Mem Inst Oswaldo Cruz. Article PubMed Google Scholar. Barletta AB, Alves LR, Silva MC, Sim S, Dimopoulos G, Liechocki S, et al.

Emerging role of lipid droplets in Aedes aegypti immune response against bacteria and dengue virus. Sci Rep. Martín-Acebes MA, Blázquez A-B, de Oya N, Escribano-Romero E, Saiz J-C.

West Nile virus replication requires fatty acid synthesis but is independent on phosphatidylinositolphosphate lipids. Merino-Ramos T, Vázquez-Calvo Á, Casas J, Sobrino F, Saiz J-C, Martín-Acebes MA. Modification of the host cell lipid metabolism induced by hypolipidemic drugs targeting the acetyl coenzyme A carboxylase impairs West Nile virus replication.

Antimicrob Agents Chemother. Heaton NS, Perera R, Berger KL, Khadka S, Lacount DJ, Kuhn RJ, et al. Dengue virus nonstructural protein 3 redistributes fatty acid synthase to sites of viral replication and increases cellular fatty acid synthesis.

Perera R, Riley C, Isaac G, Hopf-Jannasch AS, Moore RJ, Weitz KW, et al. Dengue virus infection perturbs lipid homeostasis in infected mosquito cells. Mizushima N, Levine B, Cuervo MA, Klionsky DJ.

Autophagy fights disease through cellular self-digestion. Bryant B, Raikhel AS. Programmed autophagy in the fat body of Aedes aegypti is required to maintain egg maturation cycles. Nakamoto M, Moy RH, Xu J, Bambina S, Yasunaga A, Shelly SS, et al. Virus recognition by Toll-7 activates antiviral autophagy in Drosophila.

Moy RH, Gold B, Molleston JM, Schad V, Yanger K, Salzano M-V, et al. Antiviral autophagy restricts Rift Valley fever virus infection and is conserved from flies to mammals. Shelly S, Lukinova N, Bambina S, Berman A, Cherry S.

Autophagy is an essential component of Drosophila immunity against vesicular stomatitis virus. Zhang J, Lan Y, Li MY, Lamers MM, Fusade-Boyer M, Klemm E, et al.

Flaviviruses exploit the lipid droplet protein AUP1 to trigger lipophagy and drive virus production. Article CAS Google Scholar.

Kambris Z, Cook PE, Phuc HK, Sinkins SP. Immune activation by life-shortening Wolbachia and reduced filarial competence in mosquitoes.

The researchers further identified the precise molecule—shared by many gut bacteria within that group—that unlocks the immune-protective cascade. That molecule, the researchers noted, is not difficult to isolate and could become the basis for drugs that boost antiviral immunity in humans.

The team cautions that the results remain to be confirmed in further animal studies and then replicated in humans, but the findings point to a novel strategy that could help enhance antiviral immunity in people.

The human body, like that of other mammals, is colonized by trillions of microbes—bacteria, viruses, fungi—collectively referred to as the commensal microbiota. Current estimates suggest that there are roughly as many bacterial cells as human cells in the human body, and approximately a hundred times more bacterial genes than human genes, the vast majority of which reside in the lower gastrointestinal tract.

Low-level interferon signaling that offers antiviral protection in the absence of active infection is present in all humans shortly after birth, but where and how this signaling occurs has remained unclear.

This work provides an explanation for this phenomenon, demonstrating that this protective response arises from immune cells that reside in the walls of the colon. These cells, the work shows, release protective interferons when stimulated by a surface molecule residing on the membrane of a specific gut bacterium.

In a series of experiments conducted in cells and in animals the researchers found that one of those microbes, Bacteroides fragilis , present in the majority of human guts, initiates a signaling cascade that induces immune cells in the colon to release a protein called interferon-beta, an important immune chemical that confers antiviral protection in two ways: It induces virus-infected cells to self-destruct and also stimulates other classes of immune cells to attack the virus.

This bacterial molecule stimulates an immune-signaling pathway initiated by one of the nine toll-like receptors TLR that are part of the innate immune system. The pathway is activated when proteins on the surface of immune cells recognize certain telltale molecular patterns on the surface of various infectious organisms and marshal immune defenses against these invaders through one of the nine toll-like receptor pathways.

fragilis unlocks one of these signaling pathways when its surface molecule communicates with immune cells of the colon through their TLR-4 TRIF receptors to secrete virus-repelling interferon-beta.

Because the specific surface molecule that unlocks this cascade is not unique to B. fragilis and is also present on multiple other gut bacteria of the same family, the researchers tested whether similar immune signaling could be triggered by other bacterial species carrying that molecule.

A subset of experiments in a group of mice demonstrated that membranes containing this molecule found in multiple other species of the Bacteroides bacterial family could successfully initiate similar signaling—a finding that suggest a broader immune-protective signaling common to a wide range of gut bacteria.

To determine whether B. fragilis could protect animals from infection, the researchers tested two groups of mice, one treated with antibiotics to deplete their gut microbiota and one with intact gut microbiota. Next, the researchers exposed the treated and non-treated animals to vesicular stomatitis virus VSV , an organism that infects nearly all mammals but leads to largely asymptomatic infections in humans.

Compared with mice that did not receive antibiotics and had their gut microbiota intact, antibiotic-treated animals with depleted gut microbiota were more likely to develop active infections after exposure to the virus and to have worse disease when they did get infected.

The results demonstrated the role of gut microbes in inducing protective interferon-beta signaling and in boosting natural resistance to viral infection. Interestingly, there were no differences among mice that lacked receptors for interferon-beta regardless of whether their gut microbiota was depleted.

The observations confirmed that it is precisely through interferon-beta signaling that the commensal microbiota exerts its protective effects. Finally, to investigate whether the B. fragilis surface molecule that triggers interferon signaling in cells could also modulate how animals respond to viral infection, researchers gave animals with depleted microbiotas a purified form of the molecule in their drinking water.

When, a few days later, the animals were exposed to VSV, those pretreated with the molecule had markedly milder infections and identical survival to mice with intact gut microbiota and intact immune defenses.

The findings demonstrated that supplementation with this commensal microbial molecule is sufficient to restore the protective effects of the whole microbiota in animals with depleted gut microbiota.

The work was funded by U. Department of Defense grant no. W81XWH and the Howard Hughes Medical Institute, with partial support from National Institute of Allergy and Infectious Disease grant no.

The Collagen in Skincare mechanisms of living hosts Nitric oxide levels individual cells Antiviral defense against diseases limit virus infection and replication. Individual Antiivral in all kingdoms dieeases BCAAs and immune system, including bacteria and other unicellular organisms, againstt well as BCAAs and immune system and animals ddfense evolved mechanisms to ward of virus infection and to restrict virus dissemination between and within cells. These antiviral defenses operate either through intracellular mechanisms or through the action of other cells or extracellular molecules, such as cells of the immune system and antibodies. Several theories on the origin of life have been presented. Irrespective of molecular causality, most theories eventually converge on the emergence of compartmentalized life to provide self-replicating information an advantage in a hostile environment. Paralleling emergence of the first cells, viruses likely exerted evolutionary pressure necessitating means to restrict excessive and self-annihilating replication. On one hand, Antiviral defense against diseases

Author: Yozshushicage

5 thoughts on “Antiviral defense against diseases

  1. Es ist schade, dass ich mich jetzt nicht aussprechen kann - ich beeile mich auf die Arbeit. Ich werde befreit werden - unbedingt werde ich die Meinung aussprechen.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com