Virology tidbits

Virology tidbits

Tuesday 18 October 2016

Autophagy in CSFV and ZIKV infected cells: persistence versus neurodegenerative disease

Zika Virus (ZIKV) was first isolated in 1947 from a sentinel monkey in Uganda and associated with human infection in 1954 when neutralizing antibodies were detected in the sera of residents in India, with antibodies also being found in residents from various African countries. ZIKV is mosquitoe-borne Flavivirus that is predominantly transmitted via Aedes Agypti, although sexual transmission (female to male, male to female, male to male) and transmission via blood transfusion has also been reported.
Clinically most cases are asymptomatic and symptomatic cases only present themselves with relative benign symptoms in adults, the exception being Guillain Barre Syndrome. During in the current outbreak in the Americas and –albeit only retroactively identified- in the 2013/2014 outbreak in French Polynesia, ZIKV has been identified to a causative agent of abnormal foetal brain development, leading to congenital defects, namely microcephaly, anomalies of the CNS, miscarriages and in rare cases foetal and/or neonatal death. Studies in both wt and immunodeficient mice confirmed that the infection of pregnant mice with various strains of ZIKV including strains from Asia (SZ01, FSS1305), Oceania (H/PF/2013), Brazil (Paraiba 2015), Puerto Rico (PRVABC59) and Mexico (MEX_1_7) indeed can cause abnormal development of the (foetal) brain by apoptosis of neural progenitor cells similar to representatives of the African strain, ZIKV MR766 (Uganda, 1947) and ZIKV IbH30656 (Nigeria, 1968), tested. In addition, the infection of brain organoids, neurospheres, human (foetal) Neural Progenitor Cells (h (f) NPC) and human Neural Stem Cells (NSC) with various ZIKV strains including a primary isolate from Africa, ZIKV ArB41644, demonstrated that ZIKV can indeed induce apoptosis of proliferating but probably not of mature neurons. Recently the infection of cranial neural crest cells (CNCC) with either ZIKV MR766 or ZIKV H/PF/2013 has been reported to induce high levels of cytokines that are detrimental for neurogenesis, causing bystander apoptosis of uninfected neuronal cells. Infected CNCC do not undergo apoptosis at 24 hrs p.i. but apoptosis is increased at 72 hrs p.i. confirming that both ZIKV MR766 and ZIKV H/PF/2013 can induce apoptosis of CNCC although only approx. 8% of ZIKV infected CNCC undergo apoptosis compared to 4% of mock or DENV infected CNCC, indicating that CNCC support viral infection whilst being protected against apoptosis probably by VEGF as indicated by increased secretion of VEGF which has been shown to inhibit apoptosis of human microvascular endothelial cells (HUMEC).  

In brains of wt C57BL/6 mice intracranial infected with ZIKV MR766 day 7 postnatal and analysed day 4 p.i. brain mass is reduced compared to mock infected mice by 25%, as well as showing an increase of NeuN+  and CTIP+ cells that also positive for active Caspase-3, indicating that ZIKV MR766 does not or rarely induces apoptosis in corticospinal cells, neuronal progenitor cells and astrocytes but in mature neurons in postnatal mice.  These results are in contrast with previously discussed results that indicate that in foetal mice, mature neurons are ZIKV negative and do not undergo apoptosis upon ZIKV infection. In contrast to mice infected at day 7 postnatal, mice infected at three weeks postnatal do show less severe apoptosis as measured by the presence of active Caspase-3 although similar to day 7 postnatal infected mice, widespread apoptosis of NeuN+ cells is detected by the presence active Caspase-3. When interpreting these experiments however, one has to bear in mind that ZIKV MR766 is a neurotrophic strain adapted to mice brain due to repeated intracranial passage.


Autophagy and viral infection

Autophagy is a cellular degradation pathway that involves the sequestration of cytoplasmic component such as organelles, protein aggregates or pathogens with (transient) double membrane most commonly derived from the ER, forming a phagosome that ultimately matures into the autophagosome and is degraded by lysosomal enzymes such as acidic hydrolases upon fusion with the lysosome. In general, basal autophagy is part of the turnover of RNA, proteins and organelles but can be induced in cells undergoing various forms of stress, including starvation. Both genome wide screens and large scale proteomic basic screens identified a substantial number of autophagy related regulators, including but not limited to autophagy related genes (ATG) that initiate the formation and the maturation of autophagosomes, aided by improved imaging, as well as degrading substrates such as RNA in lysosomes.
In the case of viral infections, the interactions between positive sense RNA viruses is of particular interest since autophagosomes might contribute to the decay of (viral) RNA in lysosomes by RNase T2, RNASE1 and RNASE6/RNase K6. Although it has not been determined experimentally, viral RNA might be targeted to lysosomes directly (similar to LINE 1 via processing bodies/stress granules that interact with NDP52/CALCOCO2 or p62/SQSTM-1) or as part of a complex with viral and/or cellular proteins that form a RNA-Protein Binding complex (RBP).


Figure: General outline of the autophagy pathway


More importantly however, RNA viruses such as Chikungunya Virus (CHIKV), Coronaviruses’ (CoV), Polio Virus, Measles Virus, DENV, Hepatitis C Virus (HCV) or Classical Swine Fever Virus (CSFV) can utilize autophagosomes and/or double membrane vesicles (DMV) to increase viral replication. As discussed in previous posts, in the case of CHIKV or CoV, the formation of viral replication centers (RC) is dependent on the autophagy machinery, including ATG5 and Beclin-1, whilst in others –such as HCV- autophagy is also required for exit of the mature virions as exosomes. To prevent degradation of assembled viral particles, viral proteins that inhibit the fusion of the autophagosome with the lysosome by binding Beclin-1 for instance are expressed late in the replication cycle.



Figure: Regulation of the autophagy pathway by CoV proteins as an example for the regulation of
autophagy by viral proteins


Autophagy itself can be activated via the inhibition of the Phosphatidylinositol-3 Kinase (PI3-K)-Akt-mTOR pathway, thereby inducing the phosphorylation of the ULK-1 complex and subsequently inducing the formation of the phagosome and autophagosome. This pathway is activated by the binding of growth factors such as Insulin to their respective cell surface receptor followed by the conversion of membrane bound Phosphatidylinositol-4’,5’-bisphosphate (PIP2) to Phosphatidylinositol-3’,4’,5’-trisphosphate (PIP3) thus recruiting Akt to the plasma membrane and phosphorylating Akt. Phosphorylated Akt activates the TSC1/2 complex, the GTPase activating protein (GAP) of Rheb. Activated Rheb binds and activates the mTOR-Raptor complex, inhibiting the formation of autophagosomes and thus autophagy.
Indeed, preventing the phosphorylation of Akt both at Thr 308 and Ser473 reduces the phosphorylation of mTOR at Ser2448 thus promoting autophagy whereas the ectopic expression of a constitutively active myristoylated mutant of Akt (Akt3 E17K) inhibits autophagy.


Figure: Akt inhibits the formation of autophagosomes via mTORC-1

The inhibition of autophagy in neuronal cells can induce neurodegeneration and autophagy defects –including lysosomal defects- have been implicated in Huntington Disease and ALS as well as in microcephaly and megalencephaly-polydactyly-polymicrogyria-hydrocephalus.  



CSFV and Autophagy: where autophagy meets viral persistence

Before discussing recent results concerning ZIKV and autophagy, it is worth examining the role of autophagy in cells persistently infected with CSFV and how autophagy contributes to the survival of viral infected cells.

Classical Swine Fever Virus (CSFV) is positive strand RNA virus which is classified as a Pestivirus within the Flaviviridae, causing highly virulent disease in swine characterized by a high fever, leukopenia and hemorrhages with a mortality. In infected PK-15 and 3D4/2 cells, CSFV inhibits the type I Interferon response, leading to persistent infection in the absence of apoptosis. In a recently published study, CSFV has been shown to induce the formation of LC3-II positive (mature) autophagosomes as early as 48 hrs p.i. and both the viral E2 and NS5A proteins have been shown to localise in LC3 positive vesicles as early as 24 hrs p.i. which can be inhibited by 3-Methyladenine (3-MA) as well as by downregulating the expression of either Beclin-1 or LC3-B suggesting that CSFV does induce the formation of autophagosomes as well as autophagic flux as evidenced by decreased levels of p62/SQSTM-1 in CSFV infected PK-5 and 3D4/2 cells.
Autophagy induction by CSFV however not only increases viral replication –presumably by forming viral RC- but also decreases viral induced apoptosis and decreasing mRNA levels of genes related to the type I Interferon response and IFN stimulated genes (ISG). 

CSFV induces apoptosis via activation of the intrinsic apoptotic pathway, i.e. via activation of Caspase-9 and Caspase-3, by inducing the accumulation of mitochondrial reactive oxygen species (ROS) if autophagy is inhibited in infected PK-5 and 3D4/2 cells by shBeclin1 or shLC3 which can be inhibited by either Z-VAD (a pan caspase inhibitor) or Necrostatin-1 (Nec-1/ROS scavenger). In addition to the intrinsic pathway, the extrinsic –Caspase-8 dependent- is also triggered in CSFV infected autophagy deficient cells.
CSFV induction of mitochondrial ROS not only induces the activation of apoptosis but also induces autophagy and thus promotes the clearance of damaged mitochondria via mitophagy, which is indirectly indicated by the increase of copy numbers of mitochondrial DNA in autophagy deficient PK-5 and 3D4/2 CSFV infected cells (flow cytometry using JC-1, NAO or TMRE was not performed) and/or promotes autophagy in a Nrf-2 dependent manner.


The accumulation of ROS in CFSV infected cells also induces antiviral signalling via RIG-1 and MDA-5, including increased expression of TNF/TNF-α, IFN-β/IFNB1 and FAS/TNFRSF6 in autophagy deficient CSFV infected cells, thus inducing the extrinsic pathway in an autocrine manner, which is inhibited in shDDX58, shIFIH1 and shMAVS CSFV infected autophagy deficient cells. The induction of autophagy by CSFV therefore might not only promote the clearance of damaged mitochondria but also the decreasing transcripts of ISG via RNautophagy. Similar to other positive strand RNA viruses, CSFV might also induce autophagy via the ER stress response induced by the viral NS2 and NS5A proteins which might play a role in establishing a persistent infection. In addition, the induction of NFκ-B by oxidative stress can also autophagy. If any of those pathways however contributes to viral persistence has not been investigated yet.


Figure: CSFV inhibitors apoptosis and induces autophagy via multiple pathways

Also, so far it has not been demonstrated if CSFV induced formation of ROS also induces the induction of TLR-9 by mitochondrial DNA. In this scenario, a complex of mtDNA and TLR-9 might be degraded via autophagy and thus abrogate antiviral signalling.


ZIKV and autophagy: impairment of autophagic flux linked to ZIKV induced apoptosis ?

In the case of ZIKV, the formation of autophagosome like vesicles has been observed in ZIKV MR766 infected human primary fibroblasts and keratinocytes and in the cytoplasm of C6/36 cells infected with a ZIKV isolate from a patient in Brazil although in A549 cells infected with ZIKV H/PF/2013 no LC3 positive structures have been detected.

Following the infection human foetal neural stem cells (fNSC) infected with either ZIKV MR766, ZIKV H/PF/2013 or ZIKV IbH30656 the formation of neurospheres is significantly impaired at day 7 p.i., as indicated by fewer neurospheres present which are also smaller in size, due to the induction of apoptosis as measured by TUNEL staining. Ina accordance with previously published results, ZIKV infected fNSC derived neurospheres also exhibit a reduction of cell proliferation measured by both BrdU incorporation, suggesting that ZIKV indeed does affect the proliferation of foetal neuronal precursor cells.

Similar to ZIKV MR766 infected human keratinocytes, the infection of fNSC with ZIKV MR766 or ZIKV IbH30656 induces the formation of LC3-II positive autophagosomes, without affecting autophagic flux, indicating that ZIKV infection of fNSC induces the formation of autophagosomes without affecting the formation of autolysosomes as measured by the degradation of p62/SQSTM-1. It should be noted that the infection of HeLa cells transiently transfected with a GFP/RFP-LC3 tandem plasmid with ZIKV MR766 increases GFP+/RFP+ punctae, indicating that the formation of the autolysosome might be inhibited. The discrepancy can either be due to the use of HeLa cells or it might be possible that ZIKV infection leads to the proteasomal degradation of p62/SQSTM-1 in fNSC similar to DENV-2 infected Huh-7 cells. 

In any case, treatment of ZIKV infected fNSC with Chloroquine decreases viral RNA/viral titres whilst Rapamycin treatment increases levels of both viral RNA and titers as early as 10 hrs p.i. suggesting that autophagic flux is necessary for viral replication. Based on the results described for CSFV above, autophagic flux might be necessary to prevent viral induced apoptosis and/or antiviral signalling; therefore, treatment with Chloroquine might prevent the degradation of viral RNA-TLR complexes and thus induce antiviral signalling whereas in untreated cells residual autophagy might be sufficient for inducing a partial antiviral response.  Treatment of fNSC with 3-MA inhibits viral replication, suggesting that autophagosome formation is required for viral replication, probably for the formation of viral RC. 
The ability to initiate the formation of the autophagosome is further highlighted by results that the replication of ZIKV MR766 in Atg3 -/- MEF and Atg5 -/-/MEF as well as in MEF transfected with siATG13 or siATG3 is lower when compared to wt MEF.

In DENV and HCV infected cells, the localisation of viral proteins induces the formation of viral replication complexes by utilizing the autophagy machinery; in a similar way, CoV nsp-3 and -4 have been shown to induce the formation of LC3-II positive vesicles that are derived from the ER.

In the case of ZIKV, the expression of NS4A and NS4B induces the formation of GFP-LC3 positive punctae in HeLa-GFP-LC3 cells which is more pronounced if both NS4A and NS4B are co-expressed. Similar to ZIKV MR766 infected cells, the co-expression of NS4A and NS4B increases the percentage of GFP+/RFP+ positive punctae, indicating that autophagic flux might be inhibited, although p62/SQSTM-1 levels are decreased which is probably due to proteasomal degradation of p62/SQSTM-1 (as described above). 
Based on immunofluorescence data and on data obtained from DENV NS4A and DENV NS4B, both ZIKV NS4A and NS4B are localised at the ER, suggesting that autophagy might be induced by the ER stress response.


Figure: Topology of DENV NS4A and NS4B 


As described before, the ER stress response can initiate autophagy by negatively regulating the PI-3-K/Akt pathway. Indeed, ZIKV NS4A and NS4B reduce the phosphorylation of Akt at both Thr308 and Ser473 as well as of mTOR at Ser2448 which can be reversed by the expression of a constitutively active form of Akt, Myr-HA-Akt3 E17K, in ZIKV MR766 infected fNSC or HeLa cells, suggesting that NS4A and NS4B induce the ER stress response, although it has not proven experimentally yet. 


Figure:ZIKV NS4A and NS4B inhibits PI3-K and therefore activates the formation of autophagosomes


In addition, the expression of ZIKV NS4A and NS4B decreases the proliferation of fNSC as measured by Ki67, which is more pronounced if both NS4A and NS4B are co-expressed. Since prolonged ER stress induces not only autophagy but also apoptosis, the (co-) expression of NS4A and NS4B might induce both autophagy and apoptosis although the percentage of apoptotic cells following the transfection of fNSC with ZIKV NS4A/B has not been determined.

Figure: ZIKV NS4A and NS4B decrease the expression of Ki-67 and the proliferation of fNSC

In conclusion, the infection of fNSC with ZIKV MR766, ZIKV IbH30656 and ZIKV H/PF/2013 induces apoptosis, decreased cell proliferation, smaller neurospheres and the formation of autophagosomes with impaired autophagic flux, the latter probably induced by both NS4A and NS4B protein. The notion that ZIKV MR766 inhibits the fusion of the autophagosome with the lysosome -or lysosomal maturation- and thus induces neurodegeneration by impairing the degradation of autophagic substrates is supported by findings in ZIKV MR766 infected hNPC, the expression of LAMP-2 is downregulated.


Figure: ZIKV and autophagy


If the impairment of autophagic flux however is linked to the induction of apoptosis in ZIKV infected primary neuronal cells has not been demonstrated. It might be possible that similar to CSFV infected cells, autophagy is required for abrogating antiviral signalling.
It should be noted that the infection of hNPC with ZIKV MR766 downregulates the expression of key proteins involved in the onset and progression of stress induced autophagy. Therefore, ZIKV infection of neuronal and non-neuronal cells might inhibit stress induced and selective autophagy whilst promoting the formation of viral RC. It is therefore important to analyse if the formation of viral RC is totally or only partially dependent on the ability to induce the formation of autophagosomes since viral proteins might be able to induce some of the processes required for the formation of RC independent of the autophagy machinery. 


Table: Genes that are up- or downregulated in ZIKV MR766 infected hNPC

A screen of FDA approved revealed that both Bortezomib and Ivermectin inhibit the replication of ZIKV Mex_1_7 in infected Huh-7, HeLa and/or Human Amnion Epithelial Cells. Both Bortezomib and Ivermectin induce apoptosis of cancer cells by inhibiting autophagy, indicating that in ZIKV infected (neuronal) cells autophagy might not only support viral replication by forming viral RC but also by inhibiting apoptosis at least in the early stages of viral replication. If the induction of autophagy has also consequences for the establishment of persistently infected cells, however remains to be seen.

ResearchBlogging.org







Further reading 

Pei J, Zhao M, Ye Z, Gou H, Wang J, Yi L, Dong X, Liu W, Luo Y, Liao M, & Chen J (2014). Autophagy enhances the replication of classical swine fever virus in vitro. Autophagy, 10 (1), 93-110 PMID: 24262968 

Pei J, Deng J, Ye Z, Wang J, Gou H, Liu W, Zhao M, Liao M, Yi L, & Chen J (2016). Absence of autophagy promotes apoptosis by modulating the ROS-dependent RLR signaling pathway in classical swine fever virus-infected cells. Autophagy, 12 (10), 1738-1758 PMID: 27463126 

Summerfield A, Knötig SM, & McCullough KC (1998). Lymphocyte apoptosis during classical swine fever: implication of activation-induced cell death. Journal of virology, 72 (3), 1853-61 PMID: 9499036 


Frankel LB, Lubas M, & Lund AH (2016). Emerging connections between RNA and autophagy. Autophagy, 1-21 PMID: 27715443 

He L, Zhang YM, Lin Z, Li WW, Wang J, & Li HL (2012). Classical swine fever virus NS5A protein localizes to endoplasmic reticulum and induces oxidative stress in vascular endothelial cells. Virus genes, 45 (2), 274-82 PMID: 22718084 


He L, Zhang Y, Fang Y, Liang W, Lin J, & Cheng M (2014). Classical swine fever virus induces oxidative stress in swine umbilical vein endothelial cells. BMC veterinary research, 10 PMID: 25439655 

Zhang F, Hammack C, Ogden SC, Cheng Y, Lee EM, Wen Z, Qian X, Nguyen HN, Li Y, Yao B, Xu M, Xu T, Chen L, Wang Z, Feng H, Huang WK, Yoon KJ, Shan C, Huang L, Qin Z, Christian KM, Shi PY, Xu M, Xia M, Zheng W, Wu H, Song H, Tang H, Ming GL, & Jin P (2016). Molecular signatures associated with ZIKV exposure in human cortical neural progenitors. Nucleic acids research, 44 (18), 8610-8620 PMID: 27580721 

Calvet, G., Aguiar, R., Melo, A., Sampaio, S., de Filippis, I., Fabri, A., Araujo, E., de Sequeira, P., de Mendonça, M., de Oliveira, L., Tschoeke, D., Schrago, C., Thompson, F., Brasil, P., dos Santos, F., Nogueira, R., Tanuri, A., & de Filippis, A. (2016). Detection and sequencing of Zika virus from amniotic fluid of fetuses with microcephaly in Brazil: a case study The Lancet Infectious Diseases, 16 (6), 653-660 DOI: 10.1016/S1473-3099(16)00095-5 

Cauchemez S, Besnard M, Bompard P, Dub T, Guillemette-Artur P, Eyrolle-Guignot D, Salje H, Van Kerkhove MD, Abadie V, Garel C, Fontanet A, & Mallet HP (2016). Association between Zika virus and microcephaly in French Polynesia, 2013-15: a retrospective study. Lancet (London, England), 387 (10033), 2125-32 PMID: 26993883 

Cloëtta D, Thomanetz V, Baranek C, Lustenberger RM, Lin S, Oliveri F, Atanasoski S, & Rüegg MA (2013). Inactivation of mTORC1 in the developing brain causes microcephaly and affects gliogenesis. The Journal of neuroscience : the official journal of the Society for Neuroscience, 33 (18), 7799-810 PMID: 23637172 

Cugola FR, Fernandes IR, Russo FB, Freitas BC, Dias JL, Guimarães KP, Benazzato C, Almeida N, Pignatari GC, Romero S, Polonio CM, Cunha I, Freitas CL, Brandão WN, Rossato C, Andrade DG, Faria Dde P, Garcez AT, Buchpigel CA, Braconi CT, Mendes E, Sall AA, Zanotto PM, Peron JP, Muotri AR, & Beltrão-Braga PC (2016). The Brazilian Zika virus strain causes birth defects in experimental models. Nature, 534 (7606), 267-71 PMID: 27279226 


De Carvalho NS, De Carvalho BF, Fugaça CA, Dóris B, & Biscaia ES (2016). Zika virus infection during pregnancy and microcephaly occurrence: a review of literature and Brazilian data. The Brazilian journal of infectious diseases : an official publication of the Brazilian Society of Infectious Diseases, 20 (3), 282-9 PMID: 27102780 

Arroba AI, Rodríguez-de la Rosa L, Murillo-Cuesta S, Vaquero-Villanueva L, Hurlé JM, Varela-Nieto I, & Valverde ÁM (2016). Autophagy resolves early retinal inflammation in Igf1-deficient mice. Disease models & mechanisms, 9 (9), 965-74 PMID: 27483352 

Paul P, & Münz C (2016). Autophagy and Mammalian Viruses: Roles in Immune Response, Viral Replication, and Beyond. Advances in virus research, 95, 149-95 PMID: 27112282 

McLean JE, Wudzinska A, Datan E, Quaglino D, & Zakeri Z (2011). Flavivirus NS4A-induced autophagy protects cells against death and enhances virus replication. The Journal of biological chemistry, 286 (25), 22147-59 PMID: 21511946 

Liang Q, Luo Z, Zeng J, Chen W, Foo SS, Lee SA, Ge J, Wang S, Goldman SA, Zlokovic BV, Zhao Z, & Jung JU (2016). Zika Virus NS4A and NS4B Proteins Deregulate Akt-mTOR Signaling in Human Fetal Neural Stem Cells to Inhibit Neurogenesis and Induce Autophagy. Cell stem cell PMID: 27524440 

Barrows NJ, Campos RK, Powell ST, Prasanth KR, Schott-Lerner G, Soto-Acosta R, Galarza-Muñoz G, McGrath EL, Urrabaz-Garza R, Gao J, Wu P, Menon R, Saade G, Fernandez-Salas I, Rossi SL, Vasilakis N, Routh A, Bradrick SS, & Garcia-Blanco MA (2016). A Screen of FDA-Approved Drugs for Inhibitors of Zika Virus Infection. Cell host & microbe, 20 (2), 259-70 PMID: 27476412 

Tang, H., Hammack, C., Ogden, S., Wen, Z., Qian, X., Li, Y., Yao, B., Shin, J., Zhang, F., Lee, E., Christian, K., Didier, R., Jin, P., Song, H., & Ming, G. (2016). Zika Virus Infects Human Cortical Neural Progenitors and Attenuates Their Growth Cell Stem Cell, 18 (5), 587-590 DOI: 10.1016/j.stem.2016.02.016 

Goodfellow FT, Tesla B, Simchick G, Zhao Q, Hodge T, Brindley MA, & Stice SL (2016). Zika Virus Induced Mortality and Microcephaly in Chicken Embryos. Stem cells and development PMID: 27627457 

Sir D, Kuo CF, Tian Y, Liu HM, Huang EJ, Jung JU, Machida K, & Ou JH (2012). Replication of hepatitis C virus RNA on autophagosomal membranes. The Journal of biological chemistry, 287 (22), 18036-43 PMID: 22496373 

Adams Waldorf, K., Stencel-Baerenwald, J., Kapur, R., Studholme, C., Boldenow, E., Vornhagen, J., Baldessari, A., Dighe, M., Thiel, J., Merillat, S., Armistead, B., Tisoncik-Go, J., Green, R., Davis, M., Dewey, E., Fairgrieve, M., Gatenby, J., Richards, T., Garden, G., Diamond, M., Juul, S., Grant, R., Kuller, L., Shaw, D., Ogle, J., Gough, G., Lee, W., English, C., Hevner, R., Dobyns, W., Gale, M., & Rajagopal, L. (2016). Fetal brain lesions after subcutaneous inoculation of Zika virus in a pregnant nonhuman primate Nature Medicine DOI: 10.1038/nm.4193 


Metz P, Chiramel A, Chatel-Chaix L, Alvisi G, Bankhead P, Mora-Rodriguez R, Long G, Hamacher-Brady A, Brady NR, & Bartenschlager R (2015). Dengue Virus Inhibition of Autophagic Flux and Dependency of Viral Replication on Proteasomal Degradation of the Autophagy Receptor p62. Journal of virology, 89 (15), 8026-41 PMID: 26018155 


Huang J, Li Y, Qi Y, Zhang Y, Zhang L, Wang Z, Zhang X, & Gui L (2014). Coordinated regulation of autophagy and apoptosis determines endothelial cell fate during Dengue virus type 2 infection. Molecular and cellular biochemistry, 397 (1-2), 157-65 PMID: 25138703

Ho HK, White CC, Fernandez C, Fausto N, Kavanagh TJ, Nelson SD, & Bruschi SA (2005). Nrf2 activation involves an oxidative-stress independent pathway in tetrafluoroethylcysteine-induced cytotoxicity. Toxicological sciences : an official journal of the Society of Toxicology, 86 (2), 354-64 PMID: 15901913 

Ravikumar, B., Sarkar, S., Davies, J., Futter, M., Garcia-Arencibia, M., Green-Thompson, Z., Jimenez-Sanchez, M., Korolchuk, V., Lichtenberg, M., Luo, S., Massey, D., Menzies, F., Moreau, K., Narayanan, U., Renna, M., Siddiqi, F., Underwood, B., Winslow, A., & Rubinsztein, D. (2010). Regulation of Mammalian Autophagy in Physiology and Pathophysiology Physiological Reviews, 90 (4), 1383-1435 DOI: 10.1152/physrev.00030.2009 

Menzies FM, Fleming A, & Rubinsztein DC (2015). Compromised autophagy and neurodegenerative diseases. Nature reviews. Neuroscience, 16 (6), 345-57 PMID: 25991442 

Li, Y., Chen, B., Zou, W., Wang, X., Wu, Y., Zhao, D., Sun, Y., Liu, Y., Chen, L., Miao, L., Yang, C., & Wang, X. (2016). The lysosomal membrane protein SCAV-3 maintains lysosome integrity and adult longevity The Journal of Cell Biology DOI: 10.1083/jcb.201602090 

De Leo MG, Staiano L, Vicinanza M, Luciani A, Carissimo A, Mutarelli M, Di Campli A, Polishchuk E, Di Tullio G, Morra V, Levtchenko E, Oltrabella F, Starborg T, Santoro M, di Bernardo D, Devuyst O, Lowe M, Medina DL, Ballabio A, & De Matteis MA (2016). Autophagosome-lysosome fusion triggers a lysosomal response mediated by TLR9 and controlled by OCRL. Nature cell biology, 18 (8), 839-50 PMID: 27398910 

Draganov D, Gopalakrishna-Pillai S, Chen YR, Zuckerman N, Moeller S, Wang C, Ann D, & Lee PP (2015). Modulation of P2X4/P2X7/Pannexin-1 sensitivity to extracellular ATP via Ivermectin induces a non-apoptotic and inflammatory form of cancer cell death. Scientific reports, 5 PMID: 26552848 

Selimovic D, Porzig BB, El-Khattouti A, Badura HE, Ahmad M, Ghanjati F, Santourlidis S, Haikel Y, & Hassan M (2015). Corrigendum to "Bortezomib/proteasome inhibitor triggers both apoptosis and autophagy-dependent pathways in melanoma cells". [Cell Signal. 25(1)Jan 2013 308-18]. Cellular signalling PMID: 25666930

Friday 7 October 2016

ZIKV and ocular infections


Although being first isolated in 1947 from a sentinel rhesus monkey in forests of Uganda, until 2007 Zika Virus (ZIKV) outbreaks have been sporadic, being limited to tropical regions in Africa as well as Southeast Asia and only been associated with relative mild symptoms in about 20% of infected people. Autochthonous transmission of ZIKV outside these areas was only reported in 2007 in the Federal States of Micronesia (FSM), 2013 in French Polynesia, and in 2015 in South America, spreading to the Caribbean and North Americas within a few months. Concomitant with the advent of ZIKV in the Americas, ZIKV was implicated in the onset of neurological diseases in foetuses, neonates and adults, namely microcephaly and Guillan-Barre Syndrome (GBS); subsequently, studies in animal models (both mice and monkeys), brain organoids, neural stem cells (NSC) and human neural progenitor cells (hNPC) confirmed that ZIKV can induce abnormal brain development probably by inducing apoptosis of neural progenitor -but not mature neurons- in a Caspase dependent pathway either in ZIKV infected cells or via bystander apoptosis of uninfected neural crest cells via inducing the secretion of cytokines. As discussed before, the infection of the vaginal mucosa might promote the transmission of ZIKV to the placenta and/or of Hofbauer cells that might cross the placenta and thus infect the embryo during the early stages of neural development, thus explaining data retroactively obtained from the 2013 ZIKV outbreak that indicated an increased risk for microcephaly of infants born to mothers that were infected with ZIKV in the first two trimesters. 

In addition to microcephaly however, several case reports from Brazil indicate that neonates born to ZIKV positive mothers display an array of eye malformations including blindness, intraretinal haemorrhages, chorioretinal atrophy, optic neuritis, lens sublaxation and bilateral iris colobomas, indicating that ZIKV might also affect eye development. Furthermore, in adults, ZIKV infection has been associated with conjunctivitis as well as uveitis in 10-15% of infected adults presenting themselves with  ZIKV infection.  Similar to EBOV, viral RNA can be detected in fluid samples taken from the anterior chamber of the eye, suggesting that ZIKV can either replicate in the eye or that ZIKV is at least present (but not necessarily replicating). ZIKV therefore joins other viruses that (potentially) can cause inflammation of the eye or blindness in neonates such as Human Cytomegalovirus (HCMV) which can be acquired both antenatal and intrapartum in addition to other viruses such as Adenovirus', Picornavirus', Herpes Simplex Virus 2 or Varicella Zoster Virus/Human Herpesvirus 3. So far however, an active eye infection as a result of ZIKV infection has not been reported to transmit to household contacts. More importantly however, the presence of viral RNA in the aqueous humor might indicate that following immunosuppression, ZIKV replication might resurface and thus might be horizontally transmitted via sexual contact or via mosquitoes and thus either sustain or restart a local. ZIKV  outbreak.


In order to characterize the potential of ZIKV to cause uveitis, animal models were being used. As discussed before, ZIKV does not replicate well wt C57BL/6 mice partially because the viral NS5 protein does not block mouse derived STAT2 and thus antiviral signalling. Consequently, in mice, ZIKV pathogenesis in mice is studied either in immunocompromised mice such as AG129 or Ifnar-1 -/- mice; in theory however it might also be possible to use recombinant ZIKV generated using the recently published reverse genetic systems that contain the NS5 gene of the mouse adapted ZIKV MR766 (providing that the ZIKV MR766 NS5 inhibits mouse STAT2) . In accordance with previous results which also have been discussed in previous postings, wt mice treated with Ifnar-1 -/- antibody and infected with either ZIKV H/PF/2013 or ZIKV Paraiba 2015 (isolate from Brazil) do not develop any signs of disease although high viral titres can be detected in multiple organs, including the testes of male mice, which is in contrast with Ifnar-1 -/- mice that develop severe neuroinvasive infection with decreased survival. Viral RNA in both ZIKV H/PF/2013 and ZIKV Paraiba 2015 infected mice can be detected as early as 48 hrs p.i., increasing at day 6 p.i. (H/PF/2013) and day 7 p.i. (Paraiba 2015). Also, viral RNA could be detected in tears and in lacrimal glands of ZIKV Paraiba 2015 infected mice, suggesting the presence of either infectious viral particles and/or cell debris of ZIKV infected cells due to virus induced apoptosis of infected cells.  The presence of infectious ZIKV in eyes at day 7 p.i. was confirmed by infecting immunocompromised AG129 mice eye homogenates of ZIKV Paraiba 2015 infected Ifnar-1 -/- mice, whereas both tears from day 7 p.i. nor eye homogenates from day 28 p.i. contained infectious virus, indicating that tears are not infectious and that infetious virus is not present in the eye after the acute phase of the infection. In general, eye homogenates derived from mice at day 7 p.i. , caused more severe symptoms in AG129 mice compared to ZIKV Paraiba 2015 infection, especially ocular pathology and conjunctivitis, despite similar viral titres in spleen, brain and eyes, suggesting that viral particles derived from the eye might be adapted (which at this point is speculative). One possibility to investigate if ZIKV derived from eye homogenates is adapted to cells in visual system might be to infect Ifnar-1 -/- or AG129 mice with virus grown in human retinal pigment cells (ARPE-19), retinal ganglion cells or adult retinal stem cells. Nonetheless, so far sequencing eye derived virus did not reveal substitutions except an increase single mutation in the viral NS2A gene (C to T at position 3895 or A to V), but the significance of this mutation has not been evaluated.   
As discussed in a previous post, the in utero infection of foetal mice at E4.5 via the vaginal mucosa increases the severity of foetal and neonatal brain abnormalities compared to mice infected at E8.5, confirming previous observations during the ZIKV outbreak in French Polynesia that infants born to women who got infected with ZIKV during the first trimester display a higher percentage of microcephaly. Accordingly, the offspring of Ifnar-1 -/- crossbred with Ifnar-1 +/+ mice and infected in utero  with ZIKV Paraiba 2015 at E6.5 but not E12.5 present themselves with intrauterine growth restriction (IUGR) with foetal demise and viral RNA in the brain without any (detectable) ocular defects. Viral RNA was only detected in 2 out 41 mice infected with either ZIKV H/PF/2013 or ZIKV Paraiba 2015 at day 8 post natal (or 1 week of age), indicating that ocular defects and persistence of viral RNA in the eye is indeed a rare occurrence following infection with ZIKV in utero, which is supported by epidemiological findings. In contrast, the infection of 1 week old wt mice with ZIKV Paraiba 2015 not only is lethal (in accordance with findings from the early 1950s using wt mice and the primary ZIKV MR766 isolate) but also leads to high viral RNA levels in the spleen, brain and the eye concomitant with prominent Caspase-3 dependent apoptosis of cells of the optic tract, the visual cortex and the lateral geniculate nucleus as well as other components of the visual cortex, although based on the data available it is not clear if apoptosis is also induced by "bystander apoptosis" of non-infected cells in addition to ZIKV virus infected cells. Immunodeficient AG129 infected postnatally also exhibit high viral titres at day 8 p.i. in the spleen, brain and eye irrespective if the parental or a mouse-adapted ZIKV Paraiba 2015 strain is used, suggesting that the IFN response in 1 week old wt mice is unable to inhibit viral replication irrespective of the ability of ZIKV to inhibit STAT2 signalling, which might be attributed to an immature immune system. 
In contrast to 1 week old wt/AG129 mice, adult Ifnar-1 -/- mice infected with ZIKV Paraiba 2015 present themselves with extensive apoptosis of retinal cells in the neurosensory retina as measured using TUNEL staining that detects fragmented DNA and uveitis at day 6 p.i., accompanied by infiltration of inflammatory cells into both the anterior and posterior chambers of the eye in the absence of pan-retinal damage to the fundus. Although it is not clear how ZIKV Paraiba 2015 causes uveitis, it might be possible that the viral RNA triggers an inflammatory response that not causes localised apoptosis but also triggers the inflammation of the eye via Toll-like and/or RIG-1/MDA-5 mediated signalling pathways. In any case, viral infection is cleared by day 28 p.i.. Viral RNA can be detected in all regions of the eye between 6 and 8 days p.i. with the highest levels of viral RNA in the retinal epithelium/choroid complex, in particular bipolar and ganglion cell neurons as well as the optic nerve and cornea of ZIKV infected mice as detected by FISH and qRT-PCR, indicating that ZIKV can not infect the eye but also replicates in the eye. 



In conclusion, similar to other viruses such as West Nile Virus, Hepatitis C Virus or EBOV, ZIKV can infect retinal tissue, causing ocular defects in adult as well as foetal/neonate mice. So far however further studies are needed to determine if ZIKV also replicates in retinal cells, which can be done using retinal cell lines. Also, it needs to be determined if ZIKV infection of the foetal brain downregulates the expression of genes that are required for the development of the visual system; experiments can be performed either by using foetal brain of in utero infected mice or alternatively brain organoids and/or neural stem cells. Data previously published suggest that genes encoding for proteins that are involved in the development of the visual system are differently expressed during brain development, which might explain the greater susceptibility of foetal mice infected at E6.5. Congenital ZIKV infection therefore might therefore not only target ocular tissue but also interfere with the development of the visual system which has been suggested by recent findings using SJL mice infected with ZIKV Brazil; it should be noted however that in congenital Ifnar-1 +/- foetuses derived from C57BL/6 Ifnar-1 -/- mice, no histological abnormalities are present unless they are rare and thus were not detected due to the sample size. Bystander apoptosis in non ZIKV infected cells can be induced by the secretion of cytokines by ZIKV cranial neural crest cells (CNCC). In contrast to other cell types, these cells undergo only limited apoptosis upon infection with ZIKV H/PF/2013 and secrete LIF, IL-60, PAI-1, VEGF, MCSF, TNF-α and IL-17 inducing apoptosis of co-cultured neurospheres whilst preventing viral induced apoptosis of infected CNCC in a paracrine manner. Further experiments however are needed to determine if bystander apoptosis is also affecting ocular precursor cells. In a sideline, 4 week old cerebral organoids express higher levels of VEGF compared to hNPC at week 0, so that it might be possible that ZIKV infection of 4 week old cerebral organoids even further increases the expression of VEGF in CNCC and thus prevents apoptosis of CNCC via a paracrine effect. Finally, if ZIKV replicates in retinal epithelial cells, then it might inhibit STAT3 signalling and thus lead to the degeneration of retinal epithelial cells.
Figure: Gene groups that are upregualted in 8 week old cerebral organoids compared to 4 week old
cerebral organiods









ResearchBlogging.org




































































































































































































































Further reading


Basu, R., & Tumban, E. (2016). Zika Virus on a Spreading Spree: what we now know that was unknown in the 1950’s Virology Journal, 13 (1) DOI: 10.1186/s12985-016-0623-2

Ventura CV, Maia M, Travassos SB, Martins TT, Patriota F, Nunes ME, Agra C, Torres VL, van der Linden V, Ramos RC, Rocha MÂ, Silva PS, Ventura LO, & Belfort R Jr (2016). Risk Factors Associated With the Ophthalmoscopic Findings Identified in Infants With Presumed Zika Virus Congenital Infection. JAMA ophthalmology, 134 (8), 912-8 PMID: 27228275 

Furtado JM, Espósito DL, Klein TM, Teixeira-Pinto T, & da Fonseca BA (2016). Uveitis Associated with Zika Virus Infection. The New England journal of medicine, 375 (4), 394-6 PMID: 27332784

Grant A, Ponia SS, Tripathi S, Balasubramaniam V, Miorin L, Sourisseau M, Schwarz MC, Sánchez-Seco MP, Evans MJ, Best SM, & García-Sastre A (2016). Zika Virus Targets Human STAT2 to Inhibit Type I Interferon Signaling. Cell host & microbe, 19 (6), 882-90 PMID: 27212660 

Khairallah M, Chee SP, Rathinam SR, Attia S, & Nadella V (2010). Novel infectious agents causing uveitis. International ophthalmology, 30 (5), 465-83 PMID: 19711015 

Teitelbaum BA, Newman TL, & Tresley DJ (2007). Occlusive retinal vasculitis in a patient with West Nile virus. Clinical & experimental optometry, 90 (6), 463-7 PMID: 17958570 

Goodfellow F, Tesla B, Simchick G, Hodge T, Zhao Q, Brindley MA, & Stice SL (2016). Zika Virus Induced Mortality and Microcephaly in Chicken Embryos. Stem cells and development PMID: 27627457 

Miner JJ, Sene A, Richner JM, Smith AM, Santeford A, Ban N, Weger-Lucarelli J, Manzella F, Rückert C, Govero J, Noguchi KK, Ebel GD, Diamond MS, & Apte RS (2016). Zika Virus Infection in Mice Causes Panuveitis with Shedding of Virus in Tears. Cell reports, 16 (12), 3208-18 PMID: 27612415 

Kim K, & Shresta S (2016). Neuroteratogenic Viruses and Lessons for Zika Virus Models. Trends in microbiology, 24 (8), 622-36 PMID: 27387029 

Lazear HM, Govero J, Smith AM, Platt DJ, Fernandez E, Miner JJ, & Diamond MS (2016). A Mouse Model of Zika Virus Pathogenesis. Cell host & microbe, 19 (5), 720-30 PMID: 27066744 Rossi SL, Tesh RB, Azar SR, Muruato AE, Hanley KA, Auguste AJ, Langsjoen RM, Paessler S, Vasilakis N, & Weaver SC (2016). Characterization of a Novel Murine Model to Study Zika Virus. The American journal of tropical medicine and hygiene, 94 (6), 1362-9 PMID: 27022155 

Varkey JB, Shantha JG, Crozier I, Kraft CS, Lyon GM, Mehta AK, Kumar G, Smith JR, Kainulainen MH, Whitmer S, Ströher U, Uyeki TM, Ribner BS, & Yeh S (2015). Persistence of Ebola Virus in Ocular Fluid during Convalescence. The New England journal of medicine, 372 (25), 2423-7 PMID: 25950269 

Bayless NL, Greenberg RS, Swigut T, Wysocka J, & Blish CA (2016). Zika Virus Infection Induces Cranial Neural Crest Cells to Produce Cytokines at Levels Detrimental for Neurogenesis. Cell host & microbe PMID: 27693308 

Dang J, Tiwari SK, Lichinchi G, Qin Y, Patil VS, Eroshkin AM, & Rana TM (2016). Zika Virus Depletes Neural Progenitors in Human Cerebral Organoids through Activation of the Innate Immune Receptor TLR3. Cell stem cell, 19 (2), 258-65 PMID: 27162029 

Brault JB, Khou C, Basset J, Coquand L, Fraisier V, Frenkiel MP, Goud B, Manuguerra JC, Pardigon N, & Baffet AD (2016). Comparative Analysis Between Flaviviruses Reveals Specific Neural Stem Cell Tropism for Zika Virus in the Mouse Developing Neocortex. EBioMedicine, 10, 71-6 PMID: 27453325 Patel AK, Syeda S, & Hackam AS (2013). Signal transducer and activator of transcription 3 (STAT3) signaling in retinal pigment epithelium cells. JAK-STAT, 2 (4) PMID: 24416648