key: cord-0767505-zgyfwowq authors: Harak, Christian; Lohmann, Volker title: Ultrastructure of the replication sites of positive-strand RNA viruses date: 2015-03-05 journal: Virology DOI: 10.1016/j.virol.2015.02.029 sha: 509852f6ae162ffd2d03d75f26c7d0a6ca4daef7 doc_id: 767505 cord_uid: zgyfwowq Positive strand RNA viruses replicate in the cytoplasm of infected cells and induce intracellular membranous compartments harboring the sites of viral RNA synthesis. These replication factories are supposed to concentrate the components of the replicase and to shield replication intermediates from the host cell innate immune defense. Virus induced membrane alterations are often generated in coordination with host factors and can be grouped into different morphotypes. Recent advances in conventional and electron microscopy have contributed greatly to our understanding of their biogenesis, but still many questions remain how viral proteins capture membranes and subvert host factors for their need. In this review, we will discuss different representatives of positive strand RNA viruses and their ways of hijacking cellular membranes to establish replication complexes. We will further focus on host cell factors that are critically involved in formation of these membranes and how they contribute to viral replication. Positive-strand RNA viruses replicate in the cytoplasm of an infected host cell, where they are confronted with host cell defense mechanisms and rather unfavorable conditions for genome replication. Therefore, the virus has to establish an intracellular environment that concentrates the viral proteins and allows productive replication of the viral genome, which is facilitated by induction of membranous alterations mediated by viral proteins. Host factors involved in lipid biosynthesis or vesicular trafficking are often recruited and regulated by viral proteins as well, contributing to the biogenesis of virus-induced membrane compartments. These host-derived membranes anchor the components of the replicase complex, protect the viral replication machinery from host cell immunity and allow creating a protected environment for RNA synthesis and packaging of the viral genome. Despite a great diversity between the different families of positivestrand RNA viruses and between the organelles used as origin for membrane capturing, the morphology of the replication factories shares many similarities. In general, two distinct types of membrane alterations have been identified in the last decades: the double membrane vesicle (DMV) type (Nidovirales, Picornaviridae and Hepaciviruses) and the spherule invagination type (Flaviviruses, Togaviridae, Bromoviridae, Nodaviridae). Thus, it appears that there may be common mechanisms to modulate host membranes and lipid homeostasis. In the last years, great advances have been made in imaging and visualization of subcellular structures. Techniques like electron tomography (for a technical introduction, see McIntosh et al. (2005) ) allow reconstruction of the three-dimensional architecture of viral replication complexes in nanometer-scale resolution, revealing astonishing structural details, which have not been observed in traditional transmission electron microscopy. Further developments like cryopreservation of samples and correlative light and electron microscopy (CLEM, see Sartori et al. (2007) and Lucic et al. (2013) for technical details) allow preserving the delicate ultrastructures of replication membranes and combining the advantages of fluorescently labeled proteins with the nanometer-scale resolution of electron microscopy to visualize the localization of viral or cellular proteins. In this review, we will discuss the current knowledge of the architecture of membrane rearrangements induced by different positive-strand RNA viruses. We mainly focus on studies that revealed ultrastructural details of the viral replication complexes and give some insights into the role of host factors that are hijacked to build and maintain those viral replication factories. Hepatitis C virus (HCV) is the most prominent member of the genus Hepacivirus in the family of Flaviviridae. Chronic HCV infections represent a global health burden and often lead to severe liver damage. The virus genome encompasses 9.6 kb in length and encodes for one polyprotein that is cleaved by viral and cellular proteases into 10 distinct proteins (reviewed in Bartenschlager et al. (2013) ). Virus induced membrane alteration, the so-called membranous web (MW), was initially described as accumulation of vesicles of about 85 nm in diameter embedded in a membranous matrix (Egger et al., 2002) , resembling the vesicles previously observed in poliovirus infected cells (Dales et al., 1965) . The presence of nonstructural proteins and double-stranded RNA (dsRNA) within the MW suggested these structures being the site of HCV RNA replication (Gosert et al., 2003; Ferraris et al., 2010) . The MW further was found to be closely associated with cellular lipid droplets (LDs) (Egger et al., 2002; Gosert et al., 2003; Targett-Adams et al., 2008) , which later were identified to play a major role in assembly of progeny virus (Boulant et al., 2007; Boulant et al., 2006; Salloum et al., 2013; Miyanari et al., 2007) (see also the reviews provided by Lindenbach and Rice (2013) and Lindenbach (2013) ). Detailed dissection of the MW architecture revealed that the main components are double membrane vesicles (DMVs, Fig. 1A left and top right), which probably originate as exvaginations from ER membranes and are heterogeneous in diameter (150-1000 nm). Most DMVs have two tightly opposed membrane layers, but some also exhibit two loosely surrounding lipid bilayers, which might reflect different stages of DMV formation (Ferraris et al., 2010; Romero-Brey et al., 2012) . Other studies also revealed alternative virus-induced structures like clustered vesicles, single membrane vesicles (SMVs), contiguous vesicles (Ferraris et al., 2013) or multi-membrane vesicles (MMVs) (Ferraris et al., 2010; Reiss et al., 2011) , which relevance for the HCV life cycle has not been determined so far. First DMVs are observed 16 h after infection, which correlates with increasing RNA replication, whereas MMVs occur late after around 36 h and their number further increases 48 h after infection (Romero-Brey et al., 2012) . Whether MMVs play a role in late stages of the HCV life cycle or are just a by-product of a cellular response to the DMVs is still not clear. The occurrence of MMVs might be linked to autophagic processes, but a role of autophagy in MW formation is still up to debate. MMVs might arise from self-invagination of DMVs or by double membrane tubules enwrapping DMVs (Romero-Brey et al., 2012) . In fact, upon treatment with the antiviral drug Silibinin the number of MMVs is strongly induced and RNA replication is inhibited in a genotype-specific manner (Esser-Nobis et al., 2013) . Thus, MMVs are likely inactive and do not take part in RNA replication. Currently, most evidence points to DMVs representing the structure where RNA replication takes place. Indeed, isolated DMVs revealed replicase activity in vitro . However, it still is not possible to visualize newly synthesized HCV RNA in EM micrographs, since preservation of the DMVs often is not compatible with labeling techniques and the DMVs also preclude antibodies from their inside (Romero-Brey et al., 2012) . Thus, it is still under debate whether the replication complexes reside on the surface of the DMVs or are located within the vesicles. Considering the latter hypothesis, it can be expected that the vesicles must have an opening to allow entry of nucleotides or other factors and release of viral progeny RNA. Indeed, tomographic analysis of DMVs revealed that around 10% showed a small pore that connects the inside of the vesicle with the cytosol (Figs. 1A, bottom right and 2A); however, still most of the DMVs appeared as closed structures (Romero-Brey et al., 2012) . Thus, it might be envisaged that just a small number of DMVs are actively engaged in replication at a given time point, or closed DMVs might still be active and use alternative ways to exchange metabolites, e.g. by using the nuclear transport machinery (Neufeldt et al., 2013) . Alternatively, the replication complexes might sit on the outside and the pore of some DMVs has another function or represents an epiphenomenon during membrane biogenesis. However, in vitro replicase activity in cell extracts is highly resistant to nuclease and protease treatment and only gets sensitive upon addition of detergent, suggesting that the replication complex is located within a protecting membrane structure Quinkert et al., 2005; Miyanari et al., 2003) . Although the morphology of DMVs has now been studied in great detail (Romero-Brey et al., 2012) , the different steps of DMV biogenesis are still subject to speculation. One scenario similar to DENV involves membranes invaginating into the ER leading to membrane pairing, engulfing of cytosol and subsequently formation of DMVs by further wrapping of the ER membrane ( Fig. 2A) . Alternatively, HCV proteins might induce exvagination of ER membranes resulting in a SMV that is still connected to the ER ( Fig. 2A) . In this scenario, the viral proteins would reside on the outside of the vesicles. Indeed, immunolabeling revealed that SMVs are highly enriched in viral proteins, suggesting that SMVs might represent an intermediate step in DMV biogenesis driven by viral proteins (Romero-Brey et al., 2012) . As a second step, a SMV then might undergo invagination and a DMV is formed keeping the viral proteins inside ( Fig. 2A) , which also could explain the low labeling efficiency of viral proteins at DMVs (Romero-Brey et al., 2012) . Studies of the MW are greatly facilitated by the fact that these membrane structures were not only observed in infected cells (Romero-Brey et al., 2012) or in cells harboring stably replicating subgenomic replicons (Gosert et al., 2003) , but also upon expression of viral proteins (Romero-Brey et al., 2012; Reiss et al., 2011; Berger et al., 2014) . Indeed, expression of the NS3-5B polyprotein induces the same membrane structures as observed in infection with cell culture derived virus (Romero-Brey et al., 2012). This The ER is shown in dark brown, the inner membrane of DMVs and double membrane tubules as yellow brown and their outer membrane as semi-transparent light brown. Top right: View into the lumen of a DMV connected to ER membranes. Bottom right: View of a pore (white arrow) connecting the DMV lumen to the cytosol (Romero-Brey et al., 2012) . (B) Late replication complexes of CVB3-infected cells. DMVs are shown in orange, multilamellar structures in red and parts of the neighboring ER in blue (Limpens et al., 2011) . (C) Left: Interconnected reticular network induced by DENV infection. The cytosolic face of the membrane network is shown in brown, the ER lumen in black. Right: Viral particles were found in continuous ER cisternae and are depicted in red. ER membranes are colored in light bown and inner vesicle membranes in dark brown (Welsch et al., 2009 ). (D) Left: Surface model of replication complexes induced by Kunjin virus showing ER membranes in red, ribosomes in white and viral RNA in yellow. Right: Vesicles (white) were found to be connected to each other and to ER membranes (red) (Gillespie et al., 2010) . (E) Cluster of heterogeneous DMVs induced by SARS-CoV infection. The outer DMV membrane is shown in gold, the inner membrane in silver and CMs in bronze (Knoops et al., 2008) . (F) Surface rendering of FHV replication complexes. Virally-induced spherules into the mitochondrial lumen are shown in white, mitochondrial membranes in blue. A red arrow depicts an opening of a spherule towards the cytosol (Kopek et al., 2007) . (G) 3D model of a tomographic slice of Rubella virus replication factories. Invaginated vesicles are shown in white, rigid membrane sheets in dark brown, the cytopathic vacuole in yellow, the ER in light-green and mitochondria in red (Fontana et al., 2010) . Figures were reproduced with permission from the respective journals. (SMV, top) , typically seen for Enteroviruses. The DMV is generated by an invagination event engulfing cytosol and creating a confined protected luminal space that can be connected to the cytosol by a narrow channel. Upon membrane merging, a closed DMV is created. Alternatively, DMVs can arise from exvagination of membranes into the cytosol accompanied by a secondary invagination event (bottom), as it has been suggested for HCV. (B) Replication complexes induced by DENV. An interconnected network is formed by ER membranes including SMVs that originate from invaginations into the ER. (C) Invaginated spherules as observed for BMV or FHV. A single oligomerizing viral protein (blue) is responsible for membrane alterations and forms a protein shell inside the spherules keeping them in shape. (D) Complexity of membrane alterations induced by SARS-CoV. Early stages of infection show DMVs interconnected by their outer membrane to each other as well as to convoluted membranes (CM) or ER membranes (top). Later stages of infection show membranous vesicle packets containing one or several SMVs, which can be fused to each other or exhibit membrane discontinuities (bottom) (Fontana et al., 2010) . expression model allows investigating the mechanisms of MW biogenesis also in conditions unfavorable for replication, e.g. using mutants devoid of replication or treatment with replication inhibitors. Previously, it was suggested that nonstructural protein (NS)4B is the main driver of MW biogenesis, since expression of NS4B alone was sufficient to induce MW-like structures (Egger et al., 2002) . NS4B is a highly hydrophobic integral membrane protein harboring two N-terminal amphipathic helices, four putative transmembrane domains and a highly conserved C-terminal domain. The topology of the different domains as well as oligomerization of NS4B seems to be critical for the morphology of the MW (Gouttenoire et al., 2010 (Gouttenoire et al., , 2014 Paul et al., 2011) . However, recent studies revealed that expression of NS4B alone induces only the formation of SMVs but not DMVs. In fact, all nonstructural proteins induce membrane alterations when expressed alone, mostly SMVs or single membrane tubules. Only NS5A seems to be capable of inducing DMVs upon sole expression (Romero-Brey et al., 2012) . NS5A is a multifunctional phosphoprotein harboring an N-terminal amphipathic helix associated with ER membranes, a structured domain I and two largely unstructured domains II and III (reviewed in Bartenschlager et al. (2013) ). Domain I mediates formation of different homodimeric forms and it is also suggested to form oligomers as well (Tellinghuisen et al., 2005; Love et al., 2009; Lambert et al., 2014) . Further, NS5A exists in two different phosphorylated forms, which distinct roles in HCV replication are still elusive (reviewed in Huang et al. (2007) ). To date, it is unclear if NS5A dimerization, oligomerization or the phosphorylation status might contribute to formation of the MW. Interestingly, the recently approved NS5A-specific inhibitor Daclatasvir can completely block MW formation (Berger et al., 2014) , suggesting that viral membrane alterations are indeed an attractive target for antiviral therapy. Further, NS5A interacts with a variety of host factors, which also contribute to MW architecture including PI4KIIIα (Reiss et al., 2011) and Cyclophilin A (Madan et al., 2014) . In any case, although NS5A expressed alone can induce DMV formation, a single protein is still not able to induce MW structures comparable to authentic infection. All nonstructural proteins expressed alone mediate vesicle formation, but the membrane modulating functions of all nonstructural proteins together are required for proper formation of the MW, demonstrating various complementary functions of the different proteins to generate functional replication complexes (Romero-Brey et al., 2012) . Still, we are far off understanding the molecular details of the biogenesis of the HCV replication sites. Picornaviruses comprise a diverse group of animal and human pathogens like poliovirus (PV), Hepatitis A virus (HAV), coxsackievirus (CV), foot-and-mouth-disease virus (FMDV), rhinoviruses and others. The genome of Picornaviruses comprises ca. 6.5-8 kb and encodes for a polyprotein translated by an internal ribosome entry site, which is processed by viral proteases, while the cleavage intermediates often have their own function in the viral life cycle (reviewed in Whitton et al. (2005) ). Only a few representatives of this virus family, mostly of the genus Enterovirus, have been analyzed regarding the question how Picornaviruses modulate membrane homeostasis. Today, PV is probably one of the best-studied human viruses and indeed the earliest electron microscopy images depicting membrane alterations caused by viral infection were taken from PV-infected cells (Dales et al., 1965; Kallman et al., 1958) . Later studies on the ultrastructure of PV replication complexes revealed occurrence of either SMVs (Bienz et al., 1980 (Bienz et al., , 1983 or DMVs (Schlegel et al., 1996) , but a more recent study demonstrated that indeed both structures appeared in infected cells in a time-dependent manner (Belov et al., 2012) . First membrane alterations occur 2-3 h after PV infection and were observed as elongated tubular SMVs originating from positive curvature of cellular membranes. These membrane structures were probably arising from Golgi membranes as several studies suggested (Bienz et al., 1983; Belov et al., 2012; Hsu et al., 2010) . The Golgi origin was confirmed by the sensitivity of PV replication to brefeldin A (Irurzun et al., 1992; Maynell et al., 1992) . However, fractionation analyses revealed that the replication complexes were associated with various other organelle membranes as well, arguing for different membrane sources used by the virus (Schlegel et al., 1996) . After 4 h, first DMVs of about 100-300 nm in diameter were visible and their number increased during the course of infection, while at the same time the SMVs disappeared (Belov et al., 2012) . This conversion from SMVs to DMVs probably occurs through membrane wrapping and might be due to autophagic processes (Belov et al., 2012; Limpens et al., 2011; Suhy et al., 2000) . Thus, the formation of replication vesicles seems to be a multi-step process originating from single membrane compartments to formation of DMVs. Despite the presence of dsRNA and viral proteins on both single membrane and double membrane compartments, RNA replication correlates best with the emergence of SMVs (Belov et al., 2012) . Therefore, the function of DMVs for PV is not clear, but it has been suggested that DMVs play a role in virion maturation and the non-lytic release of progeny virus (Kirkegaard and Jackson, 2005; Richards and Jackson, 2012) . Thus, PV might be able to control different steps of its life cycle by modulating the membranes of the replication complexes to either favor replication or assembly and release of virions. A three-dimensional electron tomographic study by Limpens et al. (2011) shed light on the replication complex architecture of the related Coxsackievirus B3 (CVB3) (Fig. 1B) . Similar transitions from single membrane tubules to DMVs were observed as for PV, which number greatly increased over the course of infection. About 20% of the DMVs exhibited a pore-like opening connecting the lumen to the cytosol, as it was observed for HCV (Romero-Brey et al., 2012) . The vesicles induced by CVB3 are mostly isolated compartments, which implied that the vesicles originate from exvagination of cellular compartments (Limpens et al., 2011) . Late in infection, an increasing number of DMVs were enwrapped by other membranes resulting in multilamellar vesicles occupying large areas in the cytoplasm (Fig. 1B) , reminiscent to MMVs occurring in late HCV infection (Romero-Brey et al., 2012) . In contrast to the MMVs in HCV infection, the multilamellar vesicles are enwrapped DMVs and always remain open to the cytoplasm (Limpens et al., 2011) . It still is not clear whether these structures contribute to the viral life cycle or if they are just the result of a cellular response to the DMVs. Of the seven nonstructural proteins of Enteroviruses, only three harbor hydrophobic domains (2B, 2C and 3A). 2B and 2C both have amphipathic helices that might contribute to membrane modulation (van Kuppeveld et al., 1996; Echeverri and Dasgupta, 1995; Paul et al., 1994; Teterina et al., 1997) . For the 2B protein, a model has been suggested where 2B acts as a pore integrating into Golgiderived membranes modulating membrane permeability (de Jong et al., 2003) , a function that has been described also by other groups and is linked to increased cytoplasmic calcium levels (Aldabe et al., 1997; van Kuppeveld et al., 1997) . However, a membrane modulating function has been mainly assigned to the 2BC precursor protein (Barco and Carrasco, 1995; Cho et al., 1994) , but only when coexpressed with 3A, similar membrane structures arise as observed in authentic infection (Suhy et al., 2000) . Conclusively, CVB3 seems to use only a subset of its proteins to generate the replication complexes, in contrast to HCV, which requires all of its nonstructural proteins (Romero-Brey et al., 2012). Another member of the Picornaviridae family, FMDV, causes accumulation of the majority of organelles to perinuclear regions and fragmentation of the rough ER (Monaghan et al., 2004) . Ribosomes accumulate in long chains in vicinity of the replication sites represented by SMVs and DMVs. In mid-phase and late infection, the Golgi is mostly dispersed and the number of vesicles increased, although the overall number and the proportion of DMVs were less than for other Picornaviruses. The origin of these vesicles is still not fully understood, although it is thought that they are derived from vesicles trafficking between the ER and the Golgi (Monaghan et al., 2004) , similar to the mechanism suggested for PV (Rust et al., 2001) . Despite some differences in the intracellular organization of membrane compartments between FMDV and other Picornaviruses, the same proteins (2B, 2C and 3A) seem to be of relevance. Also for FMDV, the 2BC precursor plays a major role as it is able to modulate ER membranes, in contrast to 2B or 2C expressed individually (Moffat et al., 2005) . 2B and 2C also block the secretory pathway (Moffat et al., 2007) , probably allowing the virus to use membranes involved in this process, which otherwise would be unavailable. Another recent study also demonstrated that the 3C protease is capable of blocking this pathway by inducing Golgi fragmentation (Zhou et al., 2013) . In summary, for Picornaviruses the replication most probably takes place at SMVs, which are visible early in infection and are later converted into DMVs ( Fig. 2A , top), which might fulfill functions in the late viral life cycle like assembly of viral particles. The Nidovirales are an order of positive strand RNA viruses containing, among others, the families of Coronaviridae and Arteriviridae. Coronaviruses harbor a polycistronic RNA genome with around 27-31 kb in size, which represents the largest RNA genome known to date. Besides the production of new RNA genomes from full-length negative strand RNA, they also produce subgenomic template RNAs, which are then used for production of mRNAs used for translation of structural and accessory proteins (reviewed in Masters (2006) ). Coronaviruses can infect a wide range of mammals and birds and some members impose a serious health threat to humans like the Middle East respiratory syndrome coronavirus (MERS-CoV) or the severe acute respiratory syndrom coronavirus (SARS-CoV). In infected cells, both viruses induce similar formation of paired membranes as well as ER-derived DMVs located mostly in perinuclear areas, which are also reminiscent to structures induced by mouse hepatitis coronavirus (MHV) (Goldsmith et al., 2004; Snijder et al., 2006; Knoops et al., 2008; de Wilde et al., 2013a) . First DMVs appear about 2 h after infection with a diameter of 150-300 nm. After 4 h, the number of clustered DMVs greatly increases and the vesicles are often connected to reticular membranes. Most DMVs show a tightly connected bilayer and only occasionally the membranes are loosely surrounding each other. From 3 h after infection, also CMs appear in close proximity of DMVs (David-Ferreira and Manaker, 1965; Krijnse-Locker et al., 1994) , which closely resemble the CMs observed for Flavivirus infection (Welsch et al., 2009; Westaway et al., 1997) (Fig. 1E ). The CMs induced by SARS-CoV appear largely as paired membranes having the same intermembrane distance as the DMVs (Angelini et al., 2013) and sometimes are connected to the outer membrane of DMVs and ER cisternae (Knoops et al., 2008) . Interestingly, the viral replicase proteins nsp3, nsp5 and nsp8 are located mostly in the CMs and much less in the DMVs (Goldsmith et al., 2004; Knoops et al., 2008) , similar to observations made for Kunjin virus (Westaway et al., 1997) . The DMVs in turn are largely positive for dsRNA, while the CMs are almost devoid of dsRNA (Knoops et al., 2008) , arguing for a physical segregation of viral proteins and replication intermediates. Later in infection, packets of SMVs were observed, which are surrounded by a common membrane and contained viral particles. Since many DMVs disappear during the course of infection, it is suggested that the vesicle packets might arise from merged DMVs. Electron tomographic studies confirmed that almost all DMVs are interconnected to each other by their outer membrane and are part of a continuous network with the ER (Figs. 1E and 2D). A single DMV could be connected to one or more other DMVs, to the ER or to CMs via a narrow 8 nm neck. Surprisingly, there is no connection to the cytosol, which segregates SARS-CoV from many other viruses and suggests another strategy to transport metabolites to the replication sites and to release viral RNA (Knoops et al., 2008) . The viral proteins nsp3, nsp4 and nsp6 harboring hydrophobic domains were identified to be responsible for membrane alterations (Angelini et al., 2013) , with nsp4 and nsp6 being highly conserved among the Nidovirales (Neuman et al., 2014) . Nsp3 is able to disorder and proliferate membranes. Together with nsp4, it also mediates membrane pairing. Nsp6 also has membrane proliferation activity and can induce perinuclear vesicles. All three proteins expressed together are able to induce vesicular structures very similar to those seen in authentic infection (Angelini et al., 2013) . For Arteriviruses, very similar membrane alterations have been identified consisting of an interconnected network of DMVs (Knoops et al., 2012) and nsp2 and nsp3 of the related equine arteritis virus (EAV) of the Arteriviridae family are sufficient to induce DMVs reminiscent of viral infection (Snijder et al., 2001) . Still, it is not fully understood how the viral proteins modulate host membranes, since no clear functions in membrane alterations could yet be assigned to nsp4 and nsp6 (Angelini et al., 2013) . Critical host factors involved in the biogenesis of the replication sites or the lipid composition of the replication complexes have also not been identified so far. It further remains to be determined why SARS-CoV does not require its replication vesicles being connected to the cytoplasm and why the DMVs form an interconnected network, which is an interesting and unique feature of SARS-CoV replication complexes. The genus Flavivirus includes various important human-pathogenic members like Denge virus (DENV), West Nile virus (WNV), Yellow Fever virus (YFV) or tick-borne encephalitis virus (TBEV). Flaviviruses have a capped genome of about 10-11 kb that encodes for a polyprotein processed by viral and cellular proteases to generate the mature structural and nonstructural proteins (reviewed in Chambers et al. (1990) ). DENV is an enveloped arthropod-borne pathogen and infects about 50-100 million people worldwide. Molecular aspects of DENV replication are elaborated in other reviews (Bartenschlager and Miller, 2008; Acosta et al., 2014) . First visualizations of membrane rearrangements within DENV-infected cells showed cytoplasmic vacuoles and accumulation of one or several virions in crystalloid arrays, which are surrounded by host membranes (Stohlman et al., 1975; Matsumura et al., 1971) . Later studies focused more on infected mosquito cells and visualized different steps of the viral life cycle (Hase et al., 1987; Tu et al., 1998) . Finally, the use of cryosectioning and high-pressure freezing allowed better preservation of ultrastructures and visualization of a variety of different membrane alterations (Welsch et al., 2009; Mackenzie et al., 1996a; Grief et al., 1997) . Eventually, with the use of electron tomography it was possible to image the DENV replication vesicles in three dimensions providing very detailed insights into the diverse membrane structures induced by the virus. A sophisticated study by Welsch et al. (2009) demonstrated the variety of ER-derived membrane alterations induced by DENV. Essentially, they discovered the presence of vesicle packets (VP), membrane tubules and convoluted membranes (CM). The latter ones usually were surrounded by spherical SMVs of 80-90 nm in diameter located within the lumen of the rough ER or appearing as DMV associated with ER cisternae (Fig. 1C, left) . Virus particles were observed in dilated cisternae of the rough ER in close association with the CMs and VPs (Fig. 1C, right) and could be labeled for glycoprotein E, which was not present in the VPs, suggesting distinct sites of viral RNA replication and particle assembly. Three-dimensional imaging of the DENV-induced vesicular structures using electron tomography revealed that the vesicles previously appearing as DMVs are in fact part of an interconnected network via their outer membrane (Welsch et al., 2009) (Figs. 1C and 2B) . Virion-filled ER cisternae were found to be always in near vicinity of virus-induced vesicles and very often being connected to ER structures containing these vesicles. More than half of the vesicles were connected to the outer ER membrane, which leaves a pore-like opening connecting the vesicle lumen to the cytosol. These pores have a diameter of about 11 nm and could allow both entry of metabolites required for replication and exit of viral RNA for translation or viral assembly. Further, this finding argues for an invagination event of ER membranes into the ER lumen giving rise to these vesicular structures. Additionally, Welsch et al. could demonstrate that the intraluminal virions have budded from the ER and that virions are transported to the Golgi complex by secretory vesicles originating from peripheral ER cisternae. Putative viral budding sites were identified very close to the pores of the replication vesicles, which supports the idea that the pores represent an exit site for viral RNA, which then can be efficiently packaged into viral particles (Welsch et al., 2009) . Nonstructural proteins NS2B, NS3, NS4A and NS4B as well as dsRNA were located in the VPs, suggesting that RNA replication probably takes place within these structures. Notably, only a subset of the vesicles were stained positive for dsRNA, which was located at the cytosolic side or inside of the vesicles, suggesting that only a part of the vesicles are participating in active replication at a given time point, similar to observations made for HCV (Romero-Brey et al., 2012) . The biogenesis of the membrane alterations and the function of the CMs are still unclear, though. The viral protease complex NS2B/ NS3 mainly locates to the CMs, which is thought to be the location for protein translation and polyprotein processing for the related Kunjin virus, a subtype of WNV (Westaway et al., 1997) . In expression experiments, NS4A but not NS4A-2K was able to induce CMs underlining the importance of proteolytic processing of NS4A for membrane modulation and induction of CMs. It was suggested that NS4A acts via a central peripheral domain intercalating into the luminal leaflet of the ER , probably via its membranotropic regions (Nemesio et al., 2012) . NS4B also might play a role, since it is a highly hydrophobic protein that integrates into ER membranes (Miller et al., 2006) . However, since CMs were only found in mammalian but not in mosquito cells, it is still up to debate whether they play an important role in the viral life cycle (Junjhon et al., 2014) . Also for TBEV, clear differences in membrane alterations between insect and mammalian cells were observed. A comparative study showed that infected insect cells do not show any viral particles and the extent of membrane expansion and number of vesicles was lower as compared to mammalian cells (Offerdahl et al., 2012) . These differences in membrane alteration generally might reflect certain viral strategies to propagate in the different organisms, with a specific outcome according to the steps of the viral life cycle in alternate hosts. A similar membranous network was observed for Kunjin virus, where the replication sites harboring dsRNA colocalize with markers of the trans-Golgi network (Mackenzie et al., 1999) and are closely associated with the rough ER (Gillespie et al., 2010) (Fig. 1D, left) . Similar to DENV, pores were found connecting the inner part of the vesicles to the cytosol, but also to each other (Fig. 1D, right) . The presence of dsRNA within the vesicles argued that these vesicles are the site of RNA replication (Gillespie et al., 2010; Mackenzie et al., 1996b Mackenzie et al., , 1998 . Many parallels were also observed for TBEV like the occurrence of membrane-connected vesicular structures, which partly displayed pores and seem to be Golgi-derived due to the sensitivity to brefeldin A (Lorenz et al., 2003) , a drug disrupting Golgi integrity (Fujiwara et al., 1988) . Remarkably, dsRNA was only detected in vesicles found inside the ER and it was speculated that this mechanism prevents interferon induction by shielding the viral RNA from pathogen recognition receptors (Overby et al., 2010; Pichlmair, 2007) . Electron tomography on TBEV-infected cells revealed that the vesicles originate from invaginations into the ER lumen (Miorin et al., 2013) . Again, about half of the vesicles displayed a pore connected to the cytoplasm, but although the vesicles were tightly opposed to each other, they were not interconnected in contrast to vesicles induced by WNV (Gillespie et al., 2010) . Taken together, although the different Flavivirus members show some variety in the ultrastructural morphology of their replication complexes, they share many similarities: their replication sites appear as single membrane invaginations into the ER, connected to the cytoplasm by a pore and all of them induce a system of convoluted membranes, with yet to be defined functions. However, it remains to be elucidated how these different membrane compartments are generated by the viral proteins. A protein expression system would be desired to reconstitute the membrane rearrangements seen in infection in a replication-independent setup to further study the determinants of replication complex formation. Brome mosaic virus (BMV) is a well-studied plant pathogen of the family Bromoviridae and the superfamily of alphalike-viruses. BMV harbors three genomic RNAs, each encoding for a single nonstructural protein, while the structural coat protein is translated from a subgenomic RNA . Most aspects of viral replication can be reconstituted in yeast cells greatly facilitating research of this virus (Ishikawa et al., 1997; Janda and Ahlquist, 1993) . The replication complexes induced by BMV are of the invaginated spherule type and occur on ER membranes (Restrepo-Hartwig and Ahlquist, 1999; Restrepo-Hartwig and Ahlquist, 1996; Schwartz et al., 2002) . The main driver of membrane alterations is the multi-functional viral protein 1a, which localizes to the ER and recruits the RNA polymerase 2a (Chen and Ahlquist, 2000) as well as viral RNA templates (Chen et al., 2001; . These processes are probably timely regulated, since interaction of 1a with the polymerase 2a occurs preferentially before 1a induces spherule formation, thereby efficiently recruiting the polymerase into membranous replication complexes (Chen et al., 2003; Liu et al., 2009) . A recent report demonstrated that ectopic expression of the capsid protein also induces ER-derived vesicles reminiscent of those observed in infection, but the relevance of this mechanism for the viral life cycle is not clear yet (Bamunusinghe et al., 2011) . The spherules usually are between 30 and 70 nm in diameter and are often connected to the outer ER membrane, in some cases forming an invagination connected by a neck. Similar structures also have been observed for other plant viruses (Grimley et al., 1972; Hrsel and Brcak, 1964) . The spherules can contain one or several genomic RNA intermediates, and a comparably high number of 1a molecules seems to be required to maintain those structures, while the amount of 2a RNA polymerase within the spherules is comparably low (Schwartz et al., 2002) . It is proposed that 1a forms a protein shell in the vesicle interior by its strong membrane association and self-interaction (O'Reilly et al., 1995; Kao and Ahlquist, 1992; Diaz et al., 2012) (Fig. 2C) . Thus, it is remarkable that a single protein is able to fulfill a variety of tasks including membrane association, induction of curvature and maintenance of these membranes as well as recruiting other viral proteins and RNA to the replication complexes, but so far, no three-dimensional reconstruction of the replication vesicles has been published. It will be interesting to further unravel the architecture of the BMV-induced spherules, since there appear to be many parallels between the biogenesis and architecture of BMVinduced spherules and retrovirus virion budding and genome encapsidation, suggesting that positive-strand RNA viruses, reverse-transcribing viruses and dsRNA viruses might share common ancestors (reviewed in Ahlquist (2006)). Flockhouse virus (FHV) is a non-enveloped insect virus belonging to the family of Nodaviridae. It harbors two capped genomic RNAs encoding for protein A or the coat protein, respectively, while a third subgenomic RNA is produced by protein A and encodes for two further proteins taking part in suppression of RNA silencing in host cells (reviewed in Venter and Schneemann (2008)). FHV was the first virus, which replication complexes have been revealed three-dimensionally in detail by electron tomography (Kopek et al., 2007) . FHV replicates within spherules of 50-70 nm in diameter, which, in contrast to BMV, invaginate into the outer mitochondrial membrane (Miller and Ahlquist, 2002) (Fig. 1F ), although the virus can also be productively retargeted to ER membranes (Miller et al., 2003) , demonstrating a certain structural flexibility of the replication compartments. Early in infection, affected mitochondria show compressed matrices due to the formation of spherules into the mitochondrial lumen. At later timepoints, the mitochondria suffer from dissolution of cristae and swelling of the matrices resulting in severe morphological changes (Kopek et al., 2007; Miller et al., 2001) . Similar to other viral replication vesicles, all spherules contain a pore of about 10 nm connecting the lumen of the vesicle to the cytoplasm allowing diffusion of metabolites (Kopek et al., 2007) (Fig. 1F, red arrow) . As for BMV, a single protein is required for inducing spherule formation, which is protein A that also functions as RNA polymerase. It localizes to outer mitochondria membrane and acts as an integral membrane protein (Miller and Ahlquist, 2002; Miller et al., 2001) , similar to RNA polymerases of other related viruses (Gant et al., 2014) . Similar to BMV protein 1a, it self-interacts (Dye et al., 2005) and is able to recruit RNA templates for replication (Van Wynsberghe et al., 2007) . A single spherule was quantified to contain about 100 membrane spanning protein A molecules as well as 2-4 genomic RNA intermediates (Kopek et al., 2007) . Thus, it is likely that protein A forms a shell around the spherules keeping them in their shape (Fig. 2C) . Such a high protein to RNA ratio seems not to be uncommon in viral replication, since similar observations have been made for BMV (Schwartz et al., 2002) and for HCV (Quinkert et al., 2005) . It generally seems favorable to restrict the number of genome equivalents per replication vesicle, either due to space limitations in the replication vesicles, to facilitate timely coordination of replication and assembly events by having many replication vesicles work in parallel, or to limit the presence of potential activators of host cell immunity. Interestingly, for formation of spherules, protein A has to retain its replicative function and also requires the presence of a replication-competent RNA template, arguing for active viral replication as a prerequisite for spherule formation. It is not clear yet why viral replication is required for spherule formation, but is speculated that protein A might undergo conformational changes or posttranslational modifications upon RNA replication, or that newly synthesized negative strand or dsRNA then might recruit other host factors or act as initiation site for assembly of the protein A shell (Kopek et al., 2010) . Deciphering this mechanism might clarify why for some viruses it is not possible to reconstitute membrane alterations seen in infection by expressing the viral proteins alone without an RNA template. The family of Togaviridae comprises the genera Rubivirus and Alphavirus. The only member of the genus Rubivirus is Rubella virus (RUBV), which harbors a 10 kb RNA genome containing two openreading-frames encoding the two non-structural replicase components P90 and P150 or the structural proteins capsid and envelope glycoproteins E1 and E2, respectively (Frey, 1994) . The other genus Alphavirus comprises various members like Semliki Forest virus (SFV) or Sindbis virus (SV). Alphaviruses are arthropod-borne viruses and are usually transmitted by mosquitoes between avian and mammalian hosts, where infection can lead to febrile diseases like encephalitis or arthritis (reviewed in Suhrbier et al. (2012) ). The genome comprising ca. 11.5 kb has two open-reading-frames encoding for a polyprotein precursor that is cleaved into the nonstructural proteins nsP1-nsP4 or for the structural proteins, which are expressed via a subgenomic mRNA, respectively (Kaariainen and Ahola, 2002) . All these viruses induce very similar membrane alterations in infected cells, which are designated as cytopathic vacuoles (CPV) derived from modified endosomes and lysosomes. These unique virally-induced organelles can reach very large diameters ranging from 600 to 2000 nm and contain numerous smaller invaginated spherules or vesicles of about 50 nm, as well as large vacuoles, stacked membranes and rigid membrane sheets (Fontana et al., 2007; Lee et al., 1994; Magliano et al., 1998; Froshauer et al., 1988; Grimley et al., 1968) (Fig. 1G) . The formation of RUBV CPVs is induced by the viral replicase components P90 and P150, which are located within the CPVs together with dsRNA (Fontana et al., 2007) . dsRNA also locates to the cytosolic side of the CPVs indicating that the replicase components located inside the CPVs probably have a connection to the cytosol. It has been suggested that the rigid membrane sheets might function as connector, since they reach into the cytosol and were labeled positive for dsRNA and viral proteins (Fontana et al., 2010) . These membrane sheets also enwrap the large vacuoles present in many CPVs and in some cases are connected to the periphery of the vacuole. Further, tightly packed membranes were found in some cases, which show numerous openings to the cytosol and might take part in metabolite exchange between the CPVs and the surrounding environment (Fontana et al., 2010) . CPVs also recruit other organelles like rough ER cisternae, Golgi membranes or mitochondria (Fontana et al., 2010) . Rough ER membranes connect to the CPVs either via closely apposed membranes or protein bridges, while Golgi stacks connect via small peripheral vesicles contacting the CPV membrane. Mitochondria are located in near vicinity, but do not show a direct connection to CPVs. The CPVs also are connected with the endo-lysosomal pathway as demonstrated by incorporation of BSA-gold particles (Fontana et al., 2010) , which are rapidly endocytosed by the cell after addition to the culture medium. Thus, the endolysosomal function of the membranes constituting the CPVs is still active and might contribute to the formation of the CPVs. Interestingly, although Alphaviruses induce similar structures, the requirements for CPV formation are different. Expression and cleavage of the whole P1234 polyprotein are required for replication complex formation. Biogenesis of spherules takes place at the plasma membrane (Peranen and Kaariainen, 1991; Kujala et al., 2001) , where nsp1 specifically associates to the cytosolic side of the plasma membrane via an amphipathic helix (Ahola et al., 1999; Lampio et al., 2000) . The spherules are then uptaken by the endolysosomal pathway via a mechanism dependent on phosphatidylinositol 3- Table 1 Host factors involved in replication complex biogenesis of positive-strand RNA viruses covered in this review. Sphingo-lipids Signal transmission, cell recognition (Brown and London, 2000) HCV Stimulation of polymerase activity (Hirata et al., 2012) PI4P Golgi recruitment of adapter proteins, membrane identity (Clayton et al., 2013) HCV Recruitment of OSBP or FAPP2 (Khan et al., 2014) Rhinoviruses Recruitment of OSBP (Roulin et al., 2014 ) Enteroviruses Recruitment of 3D polymerase (Hsu et al., 2010) Cholesterol Lipid rafts, detergent-resistant membranes (Silvius, 2003) WNV Immune response perturbation (Mackenzie et al., 2007) Enteroviruses Impacts replication membrane architecture (Ilnytska et al., 2013) HCV Impacts replication membrane architecture ) Saturated fatty acids Membrane component RUBV Unknown, enriched in infected cells (Williams et al., 1994) Phospho-lipids Membrane component FHV Facilitates membrane association of polymerase (Castorena et al., 2010; Stapleford et al., 2009 ) Lipid metabolism and transport PI4KIIIα PI4P synthesis at the ER and plasma membrane (Clayton et al., 2013) HCV Interacts with NS5A, impacts replication membrane architecture (Reiss et al., 2011 (Reiss et al., , 2013 , induces PI4P synthesis for recruitment of OSBP or FAPP2 (Khan et al., 2014) PI4KIIIβ PI4P synthesis at Golgi membranes (Clayton et al., 2013) Enteroviruses PI4P HCV Sphingolipid transport to replication membranes (Khan et al., 2014) Vesicle transport VAP-A Binding of SNARE proteins (Weir et al., 2001) , ER/Golgi transport, OSBP binding (Wyles et al., 2002) HCV Interacts with a specific NS5A phosphoform (Evans et al., 2004) VAP-B Similar to VAP-A, forms heterodimers with VAP-A HCV Interacts with NS5A and NS5B COPI (including ARF1, GBF1, ARFGAP1, ACBD3) Retrograde Golgi/ER transport, phospholipase D activation (Mitchell et al., 2003) Enteroviruses Blocked by 3A interaction (Wessels et al., 2006) Coronaviruses Interaction with NS4 (Oostra et al., 2007) Delivery of cytoplasmic components to the lysosome (Mizushima, 2007) PV LC3 required for replication (Taylor and Kirkegaard, 2007) Coronaviruses ATG5/12 required for replication (Prentice et al., 2004 ) DENV ATG5 required for replication (Lee et al., 2008) , manipulates lipid metabolism (Heaton and Randall, 2010) HCV ATG4B/5/12, Beclin-1 required for translation and replication (Dreux et al., 2009) , autophagosome formation by inducing LC3 lipidation (Sir et al., 2008) , ATG7 involved in particle production (Tanida et al., 2009 ), ATG5 interacts with NS5B (Guevin et al., 2010) , NS4B-induced autophagy connected with Rab5 and Vps34 (Su et al., 2011) kinase and the cytoskeleton (Spuul et al., 2010) , which then leads to the formation of the CPVs. Similar to FHV, the biogenesis of Alphavirus spherules is strictly dependent on presence of viral RNA, since expression of the viral proteins alone does not induce spherule formation (Spuul et al., 2011) . In contrast to FHV, the diameter of the invaginations of SFV is directly dependent on the size of the viral template RNA (Kallio et al., 2013) . This implies that biogenesis of the spherules is dynamic and regulated during RNA synthesis. In the first part, we described the morphology of membrane alterations induced positive strand RNA viruses and the contribution of viral proteins to their biogenesis, as far as known. However, host factors are absolutely essential to form and maintain these membranous replication complexes and our knowledge on this issue is still limited but constantly increasing. In fact, the viral replication organelles often require massive expansions of particular membrane compartments and exploit cellular machineries for vesicle formation, since viruses are limited in their genome size and simply do not have the capacities to encode for all these functions. Thus, they have to develop strategies to exploit host pathways involved in lipid synthesis and membrane curvature. However, a comprehensive overview on this topic goes beyond the scope of this review and we can just put a few spotlights on commonly emerging themes. A summary of the host factors discussed in this review is listed in Table 1 . Various cellular proteins involved in membrane curvature have been reported to play a role in biogenesis of viral replication complexes like amphiphysins for alphaviruses (Neuvonen et al., 2011) and HCV (Chao et al., 2012) , or reticulons for Picornaviruses (Tang et al., 2007) and BMV (Diaz et al., 2010) . The peptidyl-prolyl cis/trans isomerase cyclophilin A (CypA) has been shown to be essential for various steps in the replication cycle of very different viruses. However, in case of HCV, it was recently shown that CypA indeed plays a role in early events of MW biogenesis (Madan et al., 2014) , probably by interacting with NS5A, which is one of the main drivers of MW formation (Romero-Brey et al., 2012) . CypA also is involved in entry and disassembly of enterovirus particles by interacting with the VP1 protein (Qing et al., 2014) and plays an important role for replication of other viruses as well, such as HIV, HBV or influenza virus (reviewed in Zhou et al. (2012) ). For example, CypA was shown to interact with p33 and viral RNA of Tombusvirus (Kovalev and Nagy, 2013) , thereby contributing to viral replication, but also binds to the nucleocapsid of coronaviruses (Luo et al., 2004) . For the human coronaviruses NL63, also another peptidyl-prolyl cis/trans isomerase, the FK506-binding protein (FKBP) was identified to play a role in replication (Carbajo-Lozoya et al., 2014) . In contrast, the feline infectious peritonitis virus, another member of the coronavirus family, only depends on CypA, but not on FKBP (Tanaka et al., 2012) . Conclusively, the functions of peptidyl-prolyl cis/trans isomerases in viral replication seem to be very diverse between different viruses, and although it probably does not directly contribute to membrane bending, it might assist to fold and translocate viral proteins or other host factors into membranes. Different members of the Picornaviridae utilize components of the secretory pathway like the COPII- (Rust et al., 2001; Trahey et al., 2012) or COPI-machinery via Arf1-GTPase and its effector GBF1 for replication complex formation (Hsu et al., 2010; Belov et al., 2007 Belov et al., , 2008 . Different inhibitors of GBF1 have indeed been shown to inhibit enteroviral replication (van der Linden et al., 2010). HCV also has been shown to depend on ARF1, GBF1 or ARFGAP1 (Goueslain et al., 2010; Li et al., 2014; Zhang et al., 2012) , but the exact mechanism has not been clarified yet. Interestingly, ARF1 possesses an N-terminal amphipathic helix that is able to bend membranes , which renders ARF1 as potential cellular candidate taking part in replication vesicle formation, although direct evidence is lacking. Still, there are profound differences between different members of the Picornaviridae in terms of COPI-dependent replication complex biogenesis. COPI has been shown to be important for Enterovirus 71 , echovirus 11 and parechovirus 1 replication (Gazina et al., 2002) . However, while the COPI-component β-COP is specifically distributed to replication sites of echovirus 11 and partly of parechovirus 1, it is not detected in the replication membranes of the related encephalomyocarditis virus. The differences in COPIdependence are indeed reflected in the sensitivity of these viruses towards Brefeldin A Gazina et al., 2002) . SARS-CoV also interacts with COPI components and is sensitive towards Brefeldin A treatment (Knoops et al., 2010; Oostra et al., 2007; McBride et al., 2007) , but integrity of the early secretory pathway seems not to be essential for SARS-CoV to remodel ER membranes (Knoops et al., 2010) , arguing for differential roles of the COPI machinery among different viruses. Autophagy plays a role for various different viruses like PV (Taylor and Kirkegaard, 2007) , coronavirus (Prentice et al., 2004) or DENV (Lee et al., 2008) . HCV replication also relies on autophagic processes (Dreux et al., 2009; Sir et al., 2008; Tanida et al., 2009) . It is currently not clear, whether only individual factors of the autophagic pathway might be involved in formation of the MW (Romero-Brey et al., 2012; Guevin et al., 2010; Su et al., 2011) , whether DMVs are in fact autophagosomes (Sir et al., 2008) , or whether autophagy contributes to the conversion of DMVs to MMVs (Romero-Brey et al., 2012) . For a more comprehensive report on the role of autophagy in viral replication, the reader is referred to another review (Chiramel et al., 2013) . Viral infections can severely impact lipid homeostasis of the infected cell by inducing de novo biosynthesis of membranes. For almost all of the examples in this review, lipidomic profiles of infected cells have been published, often showing altered levels of sterols, sphingolipids or phospholipids (Castorena et al., 2010; Lee and Ahlquist, 2003; Perera et al., 2012; Martin-Acebes et al., 2011; Diamond et al., 2010; Tam et al., 2013; Roe et al., 2011) . For example, HCV interferes with lipogenesis pathways via the core protein or NS4B by inducing cleavage of sterol regulatory element binding proteins (SREBP), which are major transcription factors for the expression of genes involved in lipid biosynthesis (Eberle et al., 2004) . Cleavage of SREBP leads to enhanced transcription of proteins involved in lipogenesis such as fatty acid synthetase (FAS) (Waris et al., 2007; Park et al., 2009) . FAS itself interacts with the HCV polymerase NS5B and stimulates its activity (Huang et al., 2013) and also plays a role in DENV and WNV replication Martin-Acebes et al., 2011) . Also, sphingolipids have been reported to be involved in HCV replication by contributing to the formation of detergent-resistant membranes harboring the replication complexes (Hirata et al., 2012; Shi et al., 2003; Aizaki et al., 2004) . Enhanced sphingolipid synthesis was found to be beneficial for HCV replication (Hirata et al., 2012) and also stimulates polymerase activity in a genotype-specific manner (Weng et al., 2010) . Further, the replication complexes are highly enriched in cholesterol , which is probably recruited via the oxysterolbinding protein (OSBP) . Enhanced cholesterol levels at the replication sites were also observed for WNV (Mackenzie et al., 2007) and enteroviruses. For enteroviruses, the cholesterol recruitment is mediated by the clathrin-mediated endocytosis pathway, which is subverted by the virus to transport cholesterol from the plasma membrane and the extracellular environment to the replication sites (Ilnytska et al., 2013) . Overall, the requirements for distinct lipid species vary widely among different viruses, e.g. Rubella virus profits from enhanced levels of saturated fatty acids (Williams et al., 1994) , while FHV and tombusviruses require phospholipids for productive replication (Castorena et al., 2010; Stapleford et al., 2009) . It would be interesting to see whether similar vesicle morphologies of different viruses are reflected by a comparable lipid composition, but so far comparative lipidomic profiles of isolated replication membranes are lacking. Modification of lipids and the enzymes involved therein represents another way of viruses to shape the lipid environment of their replication sites. Recently, it was suggested that lipid peroxidation mediated by sphingosin kinases is a process interfering with HCV replication and that blocking these processes or alleviating the effects of lipid peroxidation in the cell stimulates RNA replication of almost all genotypes (Yamane et al., 2014) . Another interesting example of lipid-modifying enzymes contributing to viral replication involves a shared feature of Picornaviruses and HCV, which is the dependency on cellular type III phosphatidylinositol 4-kinases (PI4KIIIs) and their product PI4P. PI4P plays a role as precursor for synthesis of other phosphoinositide species and itself is involved in constituting membrane identity, signaling events and protein recruitment (reviewed in Clayton et al. (2013) ), but can also induce membrane curvature (Furse et al., 2012) . PI4Ks exists as two subtypes (II and III) each with two isoforms (α and β) located in distinct organelles. The alpha-isoform of PI4KIII (PI4KIIIα) is located at the ER and is an essential host factor for HCV replication (Reiss et al., 2011; Berger et al., 2009; Borawski et al., 2009; Tai and Salloum, 2011; Trotard et al., 2009 ). The betaisoform (PI4KIIIβ) is located at Golgi membranes (reviewed in Clayton et al. (2013) ) and plays a role for certain HCV genotypes (Reiss et al., 2011; Borawski et al., 2009 ) and for Picornaviruses (Hsu et al., 2010; Belov et al., 2007 Belov et al., , 2008 . PI4P was found in increased amounts at intracellular membranes as a consequence of viral replication in cell culture (Reiss et al., 2011; Berger et al., 2011) and in liver biopsies of HCV-infected patients (Reiss et al., 2011) . Induction of PI4P synthesis is dependent on PI4KIIIα and probably occurs due to stimulation of the kinase by NS5A, which functionally interacts with PI4KIIIα via seven amino acids in domain 1 (Reiss et al., 2013) . The kinase interacts with NS5A via different domains and only the C-terminal domains constituting the catalytic core are required for HCV replication . This virus-host interaction is not only essential for HCV replication, but also is required for PI4P accumulation and MW biogenesis (Reiss et al., 2013) . Knockdown of PI4KIIIα or interfering with PI4KIIIα-NS5A interaction results in loss of PI4P induction and in a disturbed MW structure, which appears as clustered accumulation of DMVs with reduced diameter (Reiss et al., 2011 (Reiss et al., , 2013 Berger et al., 2011) , demonstrating an important role of PI4KIIIα in the morphology of the replication complexes. The specific role of PI4P in HCV replication is not clear yet and also different degrees of colocalization between PI4P and viral proteins have been reported by independent groups (Reiss et al., 2011; Wang et al., 2014; Berger et al., 2011) . Dramatically increased PI4P levels seem not to be a prerequisite for replication (Reiss et al., 2013) , but it can be envisaged that locally increased PI4P levels play an important role in MW biogenesis. In support of that, PI4P was shown to contribute to recruitment of OSBP, which is involved in HCV replication and assembly Amako et al., 2009) . OSBP was shown to transfer cholesterol to the HCV replication sites and inhibition or knockdown resulted in a similar MW phenotype as depletion of PI4P from the replication sites by directly targeting PI4KIIIα . This supports the idea that cholesterol plays a role in DMV morphology, as it has been suggested before . Also other PI4P-binding proteins were found being involved in the HCV life cycle like the Golgi-localized protein GOLPH3 for particle assembly (Bishe et al., 2012) or FAPP2 for RNA replication. FAPP2 was suggested to bind to PI4P-enriched membranes via its PH domain and thereby transporting glycosphingolipids to the replication sites (Khan et al., 2014) . Also, a possible role of ceramide transport protein (CERT) and OSBP has been implicated for HCV particle assembly (Amako et al., 2011) . Thus, it appears that locally increased PI4P pools can serve various purposes, from inducing membrane curvature (Furse et al., 2012) , generating a lipid concentration gradient between different membranes (Mesmin et al., 2013) to recruiting lipid-binding proteins and thereby shaping the lipid composition of the replication sites Bishe et al., 2012; Khan et al., 2014) . Picornaviruses, on the other hand, seem to rely more on the PI4KIIIβ isoform, since inhibition of PI4KIIIβ leads to effective block of CVB3 and PV RNA synthesis (Hsu et al., 2010; Greninger et al., 2012; Arita et al., 2011; Sasaki et al., 2012) . This is consistent with the fact that PI4KIIIβ resides in the Golgi, where the Picornavirus replication sites mainly originate from. PI4KIIIβ is highly enriched at enteroviral replication sites and the recruitment is mediated by 3A-GBF1 interaction, which then activates ARF1 and leads to sequestration of PI4KIIIβ to viral membranes (Arita et al., 2011) . The Golgi adapter protein ACBD3 might play an additional role for recruitment of PI4KIIIβ by enteroviral protein 3A (Greninger et al., 2012) , but a recent study showed that PI4KIIIβ recruitment to CVB3 replication complexes can also occur independently of ARF1, GBF1 or ACBD3 (Dorobantu et al., 2014) . However, ACBD3 still seems to be of importance, since together with the viral proteins 3A and 3B it stimulates PI4KIIIβ activity in vitro, which might account for the accumulation of PI4P at the replication sites (Ishikawa-Sasaki et al., 2014) . PI4P then might function as adapter for the viral polymerase 3D, which preferentially associates with PI4P (Hsu et al., 2010) . Like for HCV the cholesterol-transporting function of OSBP also seems to play a role for other viruses like PV and argues for similar requirements of cholesterol at the replication membranes (Arita, 2014) and for a similar role of lipid transport proteins in composing the lipids of the replication site. Interestingly, a single point mutation in the viral protein 3A can bypass the need of CVB3 for PI4KIIIβ without affecting its virulence (Thibaut et al., 2014; van der Schaar et al., 2012) . This mutation has been identified earlier as resistance mutation against enviroxime, a small molecule drug inhibiting replication of different positive-strand RNA viruses like PV, CVB3 and rhinoviruses (Heinz and Vance, 1996) . However, it also confers cross-resistance to other inhibitors like PIK93, which efficiently targets PI4KIIIβ (Borawski et al., 2009 ), or to direct knockdown of PI4KIIIβ. This finding argues for a functional relationship between 3A and PI4P or other functions of PI4KIIIβ that can be overcome by changing a single amino acid residue. The identification of a virus mutant that circumvents the need for an essential host factor might facilitate our understanding of the role of PI4P in the viral life cycle (van der Schaar et al., 2012) . In this review, we covered some selected examples of virallyinduced membrane alterations demonstrating, on one hand, the diversity of different ultrastructures, but on the other hand, also many similarities between vesicle morphologies of related or even unrelated viruses. We certainly could only cover a small selection of positive-strand RNA viruses due to space constraints, but the reader is referred to more comprehensive reviews covering the ultrastructures of additional viruses (Romero-Brey and Miller and Krijnse-Locker, 2008) . Conclusively, immense progress in visualization of cellular ultrastructure by electron microscopy and tomography allowed researchers to explore membrane morphologies in three dimensions, revealing astonishing details of membranous networks caused by viral infections. However, we are far from understanding the viral and host factors involved in their biogenesis and it is still enigmatic, how these structures are linked to RNA synthesis. In the future, it will be important to connect microscopic techniques with elaborate mutagenesis and host factor studies to further dissect the different steps of replication complex biogenesis, and ultimately to understand the contribution of viral and cellular players taking part in these massive rearrangements of host membranes on the mechanistic level. Since lipid metabolism is tightly connected with the membrane alterations, visualization and localization of single lipid species will be as important as deciphering the specific lipid composition of isolated replication membranes. This knowledge will also help to identify novel antiviral drugs that interfere with replication complex formation, either by directly targeting a viral protein, as it has been shown for the recently approved Daclatasvir interfering with a membrane modulating function of HCV NS5A, or by targeting host factors like the COP machinery or lipid-modifying enzymes, as it is investigated for treatment of enteroviral infections. By targeting the host-virus interface it is possible to design drugs with low risk of viral resistance, but interfering with host processes also harbors the risk of severe side effects as well. It surely is desirable to develop treatments preventing the virus to use host cell processes without affecting the cellular function of those. Understanding the mechanisms behind viral replication complex biogenesis and the host factors involved will help us to pursue this direction. Revisiting dengue virus-host cell interaction: new insights into molecular and cellular virology Parallels among positive-strand RNA viruses, reversetranscribing viruses and double-stranded RNA viruses Semliki Forest virus mRNA capping enzyme requires association with anionic membrane phospholipids for activity Characterization of the hepatitis C virus RNA replication complex associated with lipid rafts Poliovirus protein 2BC increases cytosolic free calcium concentrations Role of oxysterol binding protein in hepatitis C virus infection Protein kinase D negatively regulates hepatitis C virus secretion through phosphorylation of oxysterol-binding protein and ceramide transfer protein Severe acute respiratory syndrome coronavirus nonstructural proteins 3, 4, and 6 induce double-membrane vesicles Phosphatidylinositol-4 kinase III beta and oxysterol-binding protein accumulate unesterified cholesterol on poliovirus-induced membrane structure Phosphatidylinositol 4-kinase III beta is a target of enviroxime-like compounds for antipoliovirus activity Membrane-bending mechanism of amphiphysin N-BAR domains Subcellular localization and rearrangement of endoplasmic reticulum by Brome mosaic virus capsid protein A human virus protein, poliovirus protein 2BC, induces membrane proliferation and blocks the exocytic pathway in the yeast Saccharomyces cerevisiae Molecular aspects of Dengue virus replication The molecular and structural basis of advanced antiviral therapy for hepatitis C virus infection Hijacking components of the cellular secretory pathway for replication of poliovirus RNA A critical role of a cellular membrane traffic protein in poliovirus RNA replication Complex dynamic development of poliovirus membranous replication complexes Daclatasvir-like inhibitors of NS5A block early biogenesis of hepatitis C virus-induced membranous replication factories Roles for endocytic trafficking and phosphatidylinositol 4-kinase III alpha in hepatitis C virus replication Hepatitis C virus stimulates the phosphatidylinositol 4-kinase III alpha-dependent phosphatidylinositol 4-phosphate production that is essential for its replication Kinetics and location of poliovirus macromolecular synthesis in correlation to virus-induced cytopathology Intracellular distribution of poliovirus proteins and the induction of virus-specific cytoplasmic structures Role of phosphatidylinositol 4-phosphate (PI4P) and its binding protein GOLPH3 in hepatitis C virus secretion Class III phosphatidylinositol 4-kinase alpha and beta are novel host factor regulators of hepatitis C virus replication Structural determinants that target the hepatitis C virus core protein to lipid droplets Disrupting the association of hepatitis C virus core protein with lipid droplets correlates with a loss in production of infectious virus Structure and function of sphingolipid-and cholesterol-rich membrane rafts Human coronavirus NL63 replication is cyclophilin A-dependent and inhibited by non-immunosuppressive cyclosporine A-derivatives including Alisporivir Complementary transcriptomic, lipidomic, and targeted functional genetic analyses in cultured Drosophila cells highlight the role of glycerophospholipid metabolism in Flock House virus RNA replication Flavivirus genome organization, expression, and replication Proline-serine-threonine phosphatase-interacting protein 2 (PSTPIP2), a host membrane-deforming protein, is critical for membranous web formation in hepatitis C virus replication Brome mosaic virus polymerase-like protein 2a is directed to the endoplasmic reticulum by helicase-like viral protein 1a Brome mosaic virus protein 1a recruits viral RNA2 to RNA replication through a 5 0 proximal RNA2 signal An alternate pathway for recruiting template RNA to the brome mosaic virus RNA replication complex Divergent roles of autophagy in virus infection Membrane rearrangement and vesicle induction by recombinant poliovirus 2C and 2BC in human cells Mammalian phosphatidylinositol 4-kinases as modulators of membrane trafficking and lipid signaling networks Glycosphingolipid synthesis requires FAPP2 transfer of glucosylceramide Vesicle-mediated export from the ER: COPII coat function and regulation Electron microscopic study of the formation of poliovirus An electron microscope study of the development of a mouse hepatitis virus in tissue culture cells Determinants for membrane association and permeabilization of the coxsackievirus 2B protein and the identification of the Golgi complex as the target organelle MERS-coronavirus replication induces severe in vitro cytopathology and is strongly inhibited by cyclosporin A or interferon-alpha treatment Cyclophilin inhibitors block arterivirus replication by interfering with viral RNA synthesis Temporal proteome and lipidome profiles reveal hepatitis C virus-associated reprogramming of hepatocellular metabolism and bioenergetics Membrane-shaping host reticulon proteins play crucial roles in viral RNA replication compartment formation and function Bromovirus RNA replication compartment formation requires concerted action of 1a's self-interacting RNA capping and helicase domains Recruitment of PI4KIIIbeta to coxsackievirus B3 replication organelles is independent of ACBD3, GBF1, and Arf1 The autophagy machinery is required to initiate hepatitis C virus replication In vivo self-interaction of nodavirus RNA replicase protein a revealed by fluorescence resonance energy transfer SREBP transcription factors: master regulators of lipid homeostasis Amino terminal regions of poliovirus 2C protein mediate membrane binding Expression of hepatitis C virus proteins induces distinct membrane alterations including a candidate viral replication complex Analysis of hepatitis C virus resistance to silibinin in vitro and in vivo points to a novel mechanism involving nonstructural protein 4B Phosphorylation of hepatitis C virus nonstructural protein 5A modulates its protein interactions and viral RNA replication Ultrastructural and biochemical analyses of hepatitis C virus-associated host cell membranes Sequential biogenesis of host cell membrane rearrangements induced by hepatitis C virus infection Novel replication complex architecture in rubella replicon-transfected cells Three-dimensional structure of Rubella virus factories Molecular biology of rubella virus Alphavirus RNA replicase is located on the cytoplasmic surface of endosomes and lysosomes Brefeldin A causes disassembly of the Golgi complex and accumulation of secretory proteins in the endoplasmic reticulum Lipid membrane curvature induced by distearoyl phosphatidylinositol 4-phosphate Two membraneassociated regions within the Nodamura virus RNA-dependent RNA polymerase are critical for both mitochondrial localization and RNA replication Differential requirements for COPI coats in formation of replication complexes among three genera of Picornaviridae The endoplasmic reticulum provides the membrane platform for biogenesis of the flavivirus replication complex Identification of the hepatitis C virus RNA replication complex in Huh-7 cells harboring subgenomic replicons Identification of GBF1 as a cellular factor required for hepatitis C virus RNA replication Amphipathic alphahelix AH2 is a major determinant for the oligomerization of hepatitis C virus nonstructural protein 4B Aminoterminal amphipathic alpha-helix AH1 of hepatitis C virus nonstructural protein 4B possesses a dual role in RNA replication and virus production The 3A protein from multiple picornaviruses utilizes the golgi adaptor protein ACBD3 to recruit PI4KIIIbeta Intracellular localisation of dengue-2 RNA in mosquito cell culture using electron microscopic in situ hybridisation Cytoplasmic structures associated with an arbovirus infection: loci of viral ribonucleic acid synthesis Specific membranous structures associated with the replication of group A arboviruses Autophagy protein ATG5 interacts transiently with the hepatitis C virus RNA polymerase (NS5B) early during infection Human VAP-B is involved in hepatitis C virus replication through interaction with NS5A and NS5B Mapping of functional domains of the lipid kinase phosphatidylinositol 4-kinase type III alpha involved in enzymatic activity and hepatitis C virus replication An electron and immunoelectron microscopic study of dengue-2 virus infection of cultured mosquito cells: maturation events Dengue virus-induced autophagy regulates lipid metabolism Dengue virus nonstructural protein 3 redistributes fatty acid synthase to sites of viral replication and increases cellular fatty acid synthesis Sequence determinants of 3A-mediated resistance to enviroxime in rhinoviruses and enteroviruses Self-enhancement of hepatitis C virus replication by promotion of specific sphingolipid biosynthesis Ultrastructural changes in chloroplasts and cytoplasm caused by local infection of tobacco with tobacco mosaic virus and cucumber virus 4 Viral reorganization of the secretory pathway generates distinct organelles for RNA replication Hepatitis C virus replication is modulated by the interaction of nonstructural protein NS5B and fatty acid synthase Phosphorylation of hepatitis C virus NS5A nonstructural protein: a new paradigm for phosphorylationdependent viral RNA replication? Enteroviruses harness the cellular endocytic machinery to remodel the host cell cholesterol landscape for effective viral replication Involvement of membrane traffic in the replication of poliovirus genomes: effects of brefeldin A In vivo DNA expression of functional brome mosaic virus RNA replicons in Saccharomyces cerevisiae A complex comprising phosphatidylinositol 4-kinase IIIbeta, ACBD3, and Aichi virus proteins enhances phosphatidylinositol 4-phosphate synthesis and is critical for formation of the viral replication complex RNA-dependent replication, transcription, and persistence of brome mosaic virus RNA replicons in S. cerevisiae Ultrastructural characterization and three-dimensional architecture of replication sites in dengue virus-infected mosquito cells Functions of alphavirus nonstructural proteins in RNA replication Template RNA length determines the size of replication complex spherules for Semliki Forest virus Fine structure of changes produced in cultured cells sampled at specified intervals during a single growth cycle of polio virus Identification of the domains required for direct interaction of the helicase-like and polymerase-like RNA replication proteins of brome mosaic virus Modulation of hepatitis C virus genome replication by glycosphingolipids and four-phosphate adaptor protein 2 Topology of double-membraned vesicles and the opportunity for non-lytic release of cytoplasm SARS-coronavirus replication is supported by a reticulovesicular network of modified endoplasmic reticulum Integrity of the early secretory pathway promotes, but is not required for, severe acute respiratory syndrome coronavirus RNA synthesis and virus-induced remodeling of endoplasmic reticulum membranes Ultrastructural characterization of arterivirus replication structures: reshaping the endoplasmic reticulum to accommodate viral RNA synthesis Threedimensional analysis of a viral RNA replication complex reveals a virusinduced mini-organelle Nodavirus-induced membrane rearrangement in replication complex assembly requires replicase protein A, RNA templates, and polymerase activity Cyclophilin A binds to the viral RNA and replication proteins, resulting in inhibition of tombusviral replicase assembly Characterization of the budding compartment of mouse hepatitis virus: evidence that transport from the RER to the Golgi complex requires only one vesicular transport step Biogenesis of the Semliki Forest virus RNA replication complex The crystal structure of NS5A domain 1 from genotype 1a reveals new clues to the mechanism of action for dimeric HCV inhibitors Membrane binding mechanism of an RNA virus-capping enzyme Characterization of rubella virus replication complexes using antibodies to double-stranded RNA Sar1p N-terminal helix initiates membrane curvature and completes the fission of a COPII vesicle Membrane synthesis, specific lipid requirements, and localized lipid composition changes associated with a positive-strand RNA virus RNA replication protein Autophagic machinery activated by dengue virus enhances virus replication Hepatitis C virus NS5A hijacks ARFGAP1 to maintain a phosphatidylinositol 4-phosphate-enriched microenvironment The transformation of enterovirus replication structures: a three-dimensional study of single-and double-membrane compartments Virion assembly and release The ins and outs of hepatitis C virus entry and assembly An amphipathic alpha-helix controls multiple roles of brome mosaic virus protein 1a in RNA replication complex assembly and function Intracellular assembly and secretion of recombinant subviral particles from tick-borne encephalitis virus Crystal structure of a novel dimeric form of NS5A domain I protein from hepatitis C virus Cryo-electron tomography: the challenge of doing structural biology in situ Nucleocapsid protein of SARS coronavirus tightly binds to human cyclophilin A Sphingosine kinases, sphingosine-1-phosphate and sphingolipidomics Improved membrane preservation of flavivirus-infected cells with cryosectioning Immunolocalization of the dengue virus nonstructural glycoprotein NS1 suggests a role in viral RNA replication Subcellular localization and some biochemical properties of the flavivirus Kunjin nonstructural proteins NS2A and NS4A Markers for trans-Golgi membranes and the intermediate compartment localize to induced membranes with distinct replication functions in flavivirus-infected cells Cholesterol manipulation by West Nile virus perturbs the cellular immune response Inhibition of HCV replication by cyclophilin antagonists is linked to replication fitness and occurs by inhibition of membranous web formation Rubella virus replication complexes are virus-modified lysosomes West Nile virus replication requires fatty acid synthesis but is independent on phosphatidylinositol-4-phosphate lipids The molecular biology of coronaviruses Reduction of hepatitis C virus NS5A phosphorylation through its interaction with amphiphysin II Studies on the nature of dengue viruses. V. Structure and development of dengue virus in Vero cells Inhibition of poliovirus RNA synthesis by brefeldin A The cytoplasmic tail of the severe acute respiratory syndrome coronavirus spike protein contains a novel endoplasmic reticulum retrieval signal that binds COPI and promotes interaction with membrane protein New views of cells in 3D: an introduction to electron tomography A four-step cycle driven by PI(4)P hydrolysis directs sterol/PI(4)P exchange by the ER-Golgi tether OSBP Flock house virus RNA polymerase is a transmembrane protein with amino-terminal sequences sufficient for mitochondrial localization and membrane insertion Flock house virus RNA replicates on outer mitochondrial membranes in Drosophila cells Engineered retargeting of viral RNA replication complexes to an alternative intracellular membrane Modification of intracellular membrane structures for virus replication Subcellular localization and membrane topology of the Dengue virus type 2 Non-structural protein 4B The nonstructural protein 4A of dengue virus is an integral membrane protein inducing membrane alterations in a 2K-regulated manner Three-dimensional architecture of tick-borne encephalitis virus replication sites and trafficking of the replicated RNA ADP-ribosylation factor-dependent phospholipase D activation by the M3 muscarinic receptor Hepatitis C virus non-structural proteins in the probable membranous compartment function in viral genome replication The lipid droplet is an important organelle for hepatitis C virus production Autophagy: process and function Effects of foot-and-mouth disease virus nonstructural proteins on the structure and function of the early secretory pathway: 2BC but not 3A blocks endoplasmic reticulum-to-Golgi transport Inhibition of the secretory pathway by foot-and-mouth disease virus 2BC protein is reproduced by coexpression of 2B with 2C, and the site of inhibition is determined by the subcellular location of 2C The ultrastructure of the developing replication site in foot-and-mouth disease virus-infected BHK-38 cells NS4A and NS4B proteins from dengue virus: membranotropic regions Hepatitis C virus-induced cytoplasmic organelles use the nuclear transport machinery to establish an environment conducive to virus replication Does form meet function in the coronavirus replicative organelle? SH3 domain-mediated recruitment of host cell amphiphysins by alphavirus nsP3 promotes viral RNA replication Brome mosaic virus RNA replication: revealing the role of the host in RNA virus replication Biochemical and genetic analyses of the interaction between the helicase-like and polymerase-like proteins of the brome mosaic virus A threedimensional comparison of tick-borne flavivirus infection in mammalian and tick cell lines Localization and membrane topology of coronavirus nonstructural protein 4: involvement of the early secretory pathway in replication Tick-borne encephalitis virus delays interferon induction and hides its double-stranded RNA in intracellular membrane vesicles Hepatitis C virus nonstructural 4B protein modulates sterol regulatory element-binding protein signaling via the AKT pathway Studies of a putative amphipathic helix in the N-terminus of poliovirus protein 2C Architecture and biogenesis of plus-strand RNA virus replication factories NS4B self-interaction through conserved Cterminal elements is required for the establishment of functional hepatitis C virus replication complexes Morphological and biochemical characterization of the membranous hepatitis C virus replication compartment Biogenesis of type I cytopathic vacuoles in Semliki Forest virus-infected BHK cells Dengue virus infection perturbs lipid homeostasis in infected mosquito cells Reis e Sousa C: innate recognition of viruses Coronavirus replication complex formation utilizes components of cellular autophagy Cyclophilin A associates with enterovirus-71 virus capsid and plays an essential role in viral infection as an uncoating regulator Quantitative analysis of the hepatitis C virus replication complex Recruitment and activation of a lipid kinase by hepatitis C virus NS5A is essential for integrity of the membranous replication compartment kinase III alpha regulates the phosphorylation status of hepatitis C virus NS5A Brome mosaic virus RNA replication proteins 1a and 2a colocalize and 1a independently localizes on the yeast endoplasmic reticulum Brome mosaic virus helicase-and polymerase-like proteins colocalize on the endoplasmic reticulum at sites of viral RNA synthesis Intracellular vesicle acidification promotes maturation of infectious poliovirus particles Metabolomic profile of hepatitis C virus-infected hepatocytes Membranous replication factories induced by plus-strand RNA viruses Threedimensional architecture and biogenesis of membrane structures associated with hepatitis C virus replication Rhinovirus uses a phosphatidylinositol 4-phosphate/cholesterol counter-current for the formation of replication compartments at the ER-Golgi interface Cellular COPII proteins are involved in production of the vesicles that form the poliovirus replication complex Rab18 binds to hepatitis C virus NS5A and promotes interaction between sites of viral replication and lipid droplets Correlative microscopy: bridging the gap between fluorescence light microscopy and cryo-electron tomography ACBD3-mediated recruitment of PI4KB to picornavirus RNA replication sites Cellular origin and ultrastructure of membranes induced during poliovirus infection A positive-strand RNA virus replication complex parallels form and function of retrovirus capsids Hepatitis C virus RNA replication occurs on a detergent-resistant membrane that cofractionates with caveolin-2 Role of cholesterol in lipid raft formation: lessons from lipid model systems Induction of incomplete autophagic response by hepatitis C virus via the unfolded protein response Structural and functional organization of the animal fatty acid synthase Non-structural proteins 2 and 3 interact to modify host cell membranes during the formation of the arterivirus replication complex Ultrastructure and origin of membrane vesicles associated with the severe acute respiratory syndrome coronavirus replication complex Phosphatidylinositol 3-kinase-, actin-, and microtubule-dependent transport of Semliki Forest Virus replication complexes from the plasma membrane to modified lysosomes Assembly of alphavirus replication complexes from RNA and protein components in a novel trans-replication system in mammalian cells Mitochondrion-enriched anionic phospholipids facilitate flock house virus RNA polymerase membrane association Dengue virusinduced modifications of host cell membranes Rab5 and class III phosphoinositide 3-kinase Vps34 are involved in hepatitis C virus NS4B-induced autophagy Arthritogenic alphaviruses-an overview Remodeling the endoplasmic reticulum by poliovirus infection and by individual viral proteins: an autophagy-like origin for virus-induced vesicles The role of the phosphatidylinositol 4-kinase PI4KA in hepatitis C virus-induced host membrane rearrangement Lipidomic profiling of influenza infection identifies mediators that induce and resolve inflammation Suppression of feline coronavirus replication in vitro by cyclosporin A Reticulon 3 binds the 2C protein of enterovirus 71 and is required for viral replication Knockdown of autophagy-related gene decreases the production of infectious hepatitis C virus particles Visualization of doublestranded RNA in cells supporting hepatitis C virus RNA replication Modification of cellular autophagy protein LC3 by poliovirus Structure of the zinc-binding domain of an essential component of the hepatitis C virus replicase Poliovirus 2C protein determinants of membrane binding and rearrangements in mammalian cells Fitness and virulence of a coxsackievirus mutant that can circumnavigate the need for phosphatidylinositol 4-kinase class III beta Poliovirus infection transiently increases COPII vesicle budding Kinases required in hepatitis C virus entry and replication highlighted by small interference RNA screening Ultrastructural studies on the reproductive system of male Aedes aegypti (Diptera: Culicidae) infected with dengue 2 virus Coxsackievirus mutants that can bypass host factor PI4KIIIbeta and the need for high levels of PI4P lipids for replication Differential effects of the putative GBF1 inhibitors Golgicide A and AG1478 on enterovirus replication replication protein A induces membrane association of genomic RNA Coxsackie B3 virus protein 2B contains cationic amphipathic helix that is required for viral RNA replication Coxsackievirus protein 2B modifies endoplasmic reticulum membrane and plasma membrane permeability and facilitates virus release Recent insights into the biology and biomedical applications of Flock House virus A class of membrane proteins shaping the tubular endoplasmic reticulum Oxysterol-binding protein is a phosphatidylinositol 4-kinase effector required for HCV replication membrane integrity and cholesterol trafficking COPI is required for enterovirus 71 replication The cyclophilins Brome mosaic virus 1a nucleoside triphosphatase/helicase domain plays crucial roles in recruiting RNA replication templates Hepatitis C virus induces proteolytic cleavage of sterol regulatory element binding proteins and stimulates their phosphorylation via oxidative stress VAP-A binds promiscuously to both v-and tSNAREs Composition and threedimensional architecture of the dengue virus replication and assembly sites Sphingomyelin activates hepatitis C virus RNA polymerase in a genotype-specific manner A viral protein that blocks Arf1-mediated COP-I assembly by inhibiting the guanine nucleotide exchange factor GBF1 Ultrastructure of Kunjin virus-infected cells: colocalization of NS1 and NS3 with double-stranded RNA, and of NS2B with NS3, in virus-induced membrane structures Host and virus determinants of picornavirus pathogenesis and tropism Altered membrane fatty acids of cultured human retinal pigment epithelium persistently infected with rubella virus may affect secondary cellular function Vesicle-associated membrane protein-associated protein-A (VAP-A) interacts with the oxysterol-binding protein to modify export from the endoplasmic reticulum Regulation of the hepatitis C virus RNA replicase by endogenous lipid peroxidation A novel macrophage actin-associated protein (MAYP) is tyrosine-phosphorylated following colony stimulating factor-1 stimulation ARF1 and GBF1 generate a PI4P-enriched environment supportive of hepatitis C virus replication Cyclophilin A and viral infections Foot-and-mouth disease virus 3C protease induces fragmentation of the Golgi compartment and blocks intra-Golgi transport We apologize for all the papers which we could not cite due to space limitations. Work in VLs laboratory is funded by grants from the Deutsche Forschungsgemeinschaft (LO 1556/1-2; TRR77, TPA1, FOR1202 TP3)