首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
2.
Human cytomegalovirus (HCMV) UL37 proteins traffic sequentially from the endoplasmic reticulum (ER) to the mitochondria. In transiently transfected cells, UL37 proteins traffic into the mitochondrion-associated membranes (MAM), the site of contact between the ER and mitochondria. In HCMV-infected cells, the predominant UL37 exon 1 protein, pUL37x1, trafficked into the ER, the MAM, and the mitochondria. Surprisingly, a component of the MAM calcium signaling junction complex, cytosolic Grp75, was increasingly enriched in heavy MAM from HCMV-infected cells. These studies show the first documented case of a herpesvirus protein, HCMV pUL37x1, trafficking into the MAM during permissive infection and HCMV-induced alteration of the MAM protein composition.The human cytomegalovirus (HCMV) UL37 immediate early (IE) locus expresses multiple products, including the predominant UL37 exon 1 protein, pUL37x1, also known as viral mitochondrion-localized inhibitor of apoptosis (vMIA), during lytic infection (16, 22, 24, 39, 44). The UL37 glycoprotein (gpUL37) shares UL37x1 sequences and is internally cleaved, generating pUL37NH2 and gpUL37COOH (2, 22, 25, 26). pUL37x1 is essential for the growth of HCMV in humans (17) and for the growth of primary HCMV strains (20) and strain AD169 (14, 35, 39, 49) but not strain TownevarATCC in permissive human fibroblasts (HFFs) (27).pUL37x1 induces calcium (Ca2+) efflux from the endoplasmic reticulum (ER) (39), regulates viral early gene expression (5, 10), disrupts F-actin (34, 39), recruits and inactivates Bax at the mitochondrial outer membrane (MOM) (4, 31-33), and inhibits mitochondrial serine protease at late times of infection (28).Intriguingly, HCMV UL37 proteins localize dually in the ER and in the mitochondria (2, 9, 16, 17, 24-26). In contrast to other characterized, similarly localized proteins (3, 6, 11, 23, 30, 38), dual-trafficking UL37 proteins are noncompetitive and sequential, as an uncleaved gpUL37 mutant protein is ER translocated, N-glycosylated, and then imported into the mitochondria (24, 26).Ninety-nine percent of ∼1,000 mitochondrial proteins are synthesized in the cytosol and directly imported into the mitochondria (13). However, the mitochondrial import of ER-synthesized proteins is poorly understood. One potential pathway is the use of the mitochondrion-associated membrane (MAM) as a transfer waypoint. The MAM is a specialized ER subdomain enriched in lipid-synthetic enzymes, lipid-associated proteins, such as sigma-1 receptor, and chaperones (18, 45). The MAM, the site of contact between the ER and the mitochondria, permits the translocation of membrane-bound lipids, including ceramide, between the two organelles (40). The MAM also provides enriched Ca2+ microdomains for mitochondrial signaling (15, 36, 37, 43, 48). One macromolecular MAM complex involved in efficient ER-to-mitochondrion Ca2+ transfer is comprised of ER-bound inositol 1,4,5-triphosphate receptor 3 (IP3R3), cytosolic Grp75, and a MOM-localized voltage-dependent anion channel (VDAC) (42). Another MAM-stabilizing protein complex utilizes mitofusin 2 (Mfn2) to tether ER and mitochondrial organelles together (12).HCMV UL37 proteins traffic into the MAM of transiently transfected HFFs and HeLa cells, directed by their NH2-terminal leaders (8, 47). To determine whether the MAM is targeted by UL37 proteins during infection, we fractionated HCMV-infected cells and examined pUL37x1 trafficking in microsomes, mitochondria, and the MAM throughout all temporal phases of infection. Because MAM domains physically bridge two organelles, multiple markers were employed to verify the purity and identity of the fractions (7, 8, 19, 46, 47).(These studies were performed in part by Chad Williamson in partial fulfillment of his doctoral studies in the Biochemistry and Molecular Genetics Program at George Washington Institute of Biomedical Sciences.)HFFs and life-extended (LE)-HFFs were grown and not infected or infected with HCMV (strain AD169) at a multiplicity of 3 PFU/cell as previously described (8, 26, 47). Heavy (6,300 × g) and light (100,000 × g) MAM fractions, mitochondria, and microsomes were isolated at various times of infection and quantified as described previously (7, 8, 47). Ten- or 20-μg amounts of total lysate or of subcellular fractions were resolved by SDS-PAGE in 4 to 12% Bis-Tris NuPage gels (Invitrogen) and examined by Western analyses (7, 8, 26). Twenty-microgram amounts of the fractions were not treated or treated with proteinase K (3 μg) for 20 min on ice, resolved by SDS-PAGE, and probed by Western analysis. The blots were probed with rabbit anti-UL37x1 antiserum (DC35), goat anti-dolichyl phosphate mannose synthase 1 (DPM1), goat anti-COX2 (both from Santa Cruz Biotechnology), mouse anti-Grp75 (StressGen Biotechnologies), and the corresponding horseradish peroxidase-conjugated secondary antibodies (8, 47). Reactive proteins were detected by enhanced chemiluminescence (ECL) reagents (Pierce), and images were digitized as described previously (26, 47).  相似文献   

3.
Microsporidia are a group of highly adapted obligate intracellular parasites that are now recognized as close relatives of fungi. Their adaptation to parasitism has resulted in broad and severe reduction at (i) a genomic level by extensive gene loss, gene compaction, and gene shortening; (ii) a biochemical level with the loss of much basic metabolism; and (iii) a cellular level, resulting in lost or cryptic organelles. Consistent with this trend, the mitochondrion is severely reduced, lacking ATP synthesis and other typical functions and apparently containing only a fraction of the proteins of canonical mitochondria. We have investigated the mitochondrial protein import apparatus of this reduced organelle in the microsporidian Encephalitozoon cuniculi and find evidence of reduced and modified machinery. Notably, a putative outer membrane receptor, Tom70, is reduced in length but maintains a conserved structure chiefly consisting of tetratricopeptide repeats. When expressed in Saccharomyces cerevisiae, EcTom70 inserts with the correct topology into the outer membrane of mitochondria but is unable to complement the growth defects of Tom70-deficient yeast. We have scanned genomic data using hidden Markov models for other homologues of import machinery proteins and find evidence of severe reduction of this system.Microsporidia are a eukaryotic group highly adapted as obligate intracellular parasites (31, 50). They infect a diverse range of vertebrate and invertebrate animal hosts. In humans they are the cause of a number of diseases (e.g., gastroenteritis, encephalitis, and hepatitis), having their greatest impact on immunocompromised individuals, notably in children with human immunodeficiency virus (14, 31). Microsporidia are most closely related to fungi, although their high level of specialization as intracellular parasites obscured this relationship for a long time (18, 25, 30). Gene phylogenies now firmly connect these two groups, although it remains uncertain whether microsporidia are sisters to the fungi or represent a lineage derived from within fungal diversity (21, 28).A clear adaptive response to parasitism in microsporidia has been a reduction in cellular complexity. This was first recognized at an ultrastructural level with the apparent lack of peroxisomes, flagella, stacked Golgi bodies, and mitochondria (31). This reductive evolution is mirrored at a genomic level, with microsporidia containing the smallest eukaryotic genomes described to date (28, 29). The complete genomic sequence from the human microsporidian parasite Encephalitozoon cuniculi reveals a genome of only ∼2.9 Mb containing approximately 2,000 genes, in contrast to the 6,000 genes found in the genome of the model fungus Saccharomyces cerevisiae. The minimal genome of E. cuniculi has been achieved through three mechanisms in concert: (i) gene loss, resulting in broad loss of biochemical pathways and capabilities, including much basic energy metabolism and numerous anabolic pathways; (ii) gene compaction with an average intergenic space of ∼130 bp; and (iii) gene shortening, with E. cuniculi genes being on average 14% shorter than their homologues in fungi such as S. cerevisiae (28, 45). Thus, microsporidian evolution has apparently been shaped by a very strong trend to eliminate superfluous molecular and biochemical complexity.Despite earlier suppositions that microsporidia lacked mitochondria, genome and expressed sequence tag data from microsporidia suggested the presence of several proteins typically targeted to this organelle (3, 19, 20, 24, 28, 38). Immunolocalization of a mitochondrial Hsp70 to small double membrane-bound organelles in Trachipleistophora hominis provided strong evidence for the existence of a mitochondrion in microsporidia, albeit a simplified organelle that lacks cisternae (48). Annotation of genomic data from E. cuniculi provided compelling matches for only 22 proteins implicated in mitochondrial function, suggesting that the metabolism of this relict mitochondrion (or mitosome) is also significantly reduced compared to that of canonical mitochondria (28). Further, no mitochondrial genome has been retained; thus, biogenesis of this organelle is wholly dependent on nucleus-encoded proteins. Based on these 22 proteins, a major role for the mitosome is iron-sulfur cluster assembly (22, 28). No genes have been found for ATP synthesis via oxidative phosphorylation, suggesting loss of this activity in mitosomes (28, 46). While it is likely that further mitosome-targeted proteins will be identified, it is clear that compared to mitochondria from fungal relatives, which are known to import ∼1,000 proteins (40, 44), microsporidian mitosomes represent organelles with highly reduced proteomes, a feature consistent with other traits of cellular reduction.The highly reduced state of the microsporidian mitosome, requiring only a fraction of the protein diversity of other mitochondria, presents an interesting case for studying organelle biogenesis—particularly the machinery for protein import of nucleus-encoded proteins. Mitochondrial protein import has been best characterized in fungi, and in these systems most proteins are imported via four major import complexes: a TOM (translocase of the outer mitochondrial membrane), a SAM (sorting and assembly machinery), and one of two TIMs (translocase of the inner mitochondrial membrane), TIM23 or TIM22 (see Fig. Fig.5A)5A) (5, 36). These complexes are broadly conserved throughout fungi as well as animals (15). Mitochondrial proteins can take one of several routes to the mitochondrion via this apparatus (5, 36). Broadly, soluble matrix proteins are recognized at the TOM complex by the receptor protein Tom20 through the binding of N-terminal presequences with characteristic features (1, 5, 7, 8, 36). These proteins are passed through the pore protein Tom40 of the TOM to the TIM23 complex and then driven into the mitochondrial matrix by way of the presequence translocase-associated motor (PAM) complex, where their presequences are subsequently removed. Some membrane proteins can also be released into the inner membrane from the TIM23 complex. Mitochondrial proteins that apparently lack such an extension, notably including many of the membrane proteins, are recognized by internal sequence elements. Tom70 has a greater role in recognizing these internal signals and thus the import of hydrophobic proteins (4, 11, 32, 39, 47). Such hydrophobic proteins are often bound by cytosolic molecular chaperones (Hsp70 and/or Hsp90) en route to the mitochondrion, and Tom70 is known to independently bind to both the chaperone and the substrate protein (7, 23, 33, 52). While a measure of substrate overlap between Tom20 and Tom70 occurs, the division of responsibility between these two receptors has likely evolved in response to the wide range of substrate proteins that must be imported into mitochondria and the need to handle this complexity.Open in a separate windowFIG. 5.Schematics of the protein import machinery and pathways in yeast mitochondria (A) and E. cuniculi mitosome (B) based on identified homologues of the general fungal/animal pathways. Protein components of the yeast system were all represented by HMMs used to search the microsporidian genomic data and represent the major presequence-dependent and presequence-independent pathways. Homologues identified in E. cuniculi indicate a severely reduced import apparatus utilizing elements of the presequence-independent pathway.For microsporidia little is known of the protein import apparatus for their relict mitochondrion, the mitosome. Has the very reduced organelle proteome, in concert with a genome-wide trend of the loss of redundant or superfluous genes, resulted in a smaller and/or derived import apparatus? In this study we have investigated the microsporidian mitosome protein import apparatus from E. cuniculi in order to evaluate how this apparatus has responded to the reduction in the number of proteins required to be imported and the overall radical reduction in the number and size of proteins encoded in the nuclear genome. A putative homologue of the outer membrane receptor protein Tom70 is of particular interest as the only receptor for the TOM complex and, given the known structure of Tom70 proteins, provides a highly informative example of how proteins can be shortened in the course of genome reduction.  相似文献   

4.
Deformylases are metalloproteases in bacteria, plants, and humans that remove the N-formyl-methionine off peptides in vitro. The human homolog of peptide deformylase (HsPDF) resides in the mitochondria, along with its putative formylated substrates; however, the cellular function of HsPDF remains elusive. Here we report on the function of HsPDF in mitochondrial translation and oxidative phosphorylation complex biogenesis. Functional HsPDF appears to be necessary for the accumulation of mitochondrial DNA-encoded proteins and assembly of new respiratory complexes containing these proteins. Consequently, inhibition of HsPDF reduces respiratory function and cellular ATP levels, causing dependence on aerobic glycolysis for cell survival. A series of structurally different HsPDF inhibitors and control peptidase inhibitors confirmed that inhibition of HsPDF decreases mtDNA-encoded protein accumulation. Therefore, HsPDF appears to have a role in maintenance of mitochondrial respiratory function, and this function is analogous to that of chloroplast PDF.The human mitochondrial protein peptide deformylase, HsPDF, is a metalloprotease that removes the formyl moiety on the methionine of N-formyl-methionine peptide substrates in an enzymatic assay (24, 35). Despite the slow kinetic properties of HsPDF in an in vitro deformylation assay (24, 29, 35), we have shown that small interfering RNA (siRNA) interference of HsPDF decreases human cancer cell proliferation. Similarly, pharmacologic inhibition with the PDF antibiotic inhibitor actinonin and its analogs results in mitochondrial membrane depolarization and promotes cell death or proliferation arrest in a wide variety of cancer cell lines (18, 25). However, the cellular function of HsPDF remains elusive, and others have proposed that it has none (29). In bacteria, deformylation of nascent peptides is necessary for removal of the N-terminal methionine (36) and posttranslational processing of at least a subset of proteins that contribute to cell growth and viability (28). Prokaryotic PDF thus fulfills a role in cotranslational processing (7) and in protein degradation (41).In mammals, N-terminal formylation of proteins is only known to occur during mitochondrial translation initiation, as in prokaryotic protein translation (6). In contrast to bacteria, where the entire proteome is formylated for translation initiation, formylation in eukaryotes is limited to the 13 mitochondrial DNA (mtDNA)-encoded proteins. Formylation is important for mitochondrial translation, because formyl-Met-tRNA, but not Met-tRNA, is recognized by initiation factor 2 as the initiator tRNA (26, 37, 39). Therefore, the participation of HsPDF in protein post- or cotranslational processing can be narrowed down to these mitochondrial translation products.Despite the current understanding of the function of formyl-methionine in the initiation of protein synthesis in mammalian mitochondria (38, 39), the functional relevance of the downstream processing of nascent mitochondrial translation products has remained unexplored. Furthermore, it has been assumed that human mitochondria-encoded proteins, like those of bovine origin, are generally not deformylated after synthesis (45).The mammalian mitochondrial genome-encoded proteins are all subunits of four of the five oxidative phosphorylation respiratory chain enzyme complexes (I, III, IV, and V) (2, 40, 42). Respiratory complexes are comprised of multiple proteins. With the exception of complex II, which is comprised entirely of nuclear DNA-encoded subunits, all other complexes include both nuclear and mitochondrial DNA-encoded proteins. Synthesis of key mtDNA-encoded protein subunits, and the assembly of these proteins with multiple nuclear-encoded subunits within the mitochondria, is necessary for the function of each individual complex (16, 30, 44). Moreover, a functional interdependence among stably assembled respiratory complexes has been demonstrated (1). Mutations in human mtDNA that affect protein-coding regions or nuclear DNA mutations that affect expression of respiratory complex subunits cause disease (13), including Parkinson''s disease, for example, in which decreased respiratory function and compromised cell viability have been demonstrated (5, 21, 23). Therefore, the importance of properly assembled mitochondrial respiratory complexes suggests that their disruption, by inhibition of mtDNA-encoded protein processing, could have significant effects on cellular function.We hypothesized that HsPDF-mediated processing of mtDNA-encoded proteins is necessary for proper function of the respiratory chain complexes. To determine how the human deformylase activity contributes to cellular function, we used pharmacologic inhibition of HsPDF activity with the hydroxamic acid peptidomimetic inhibitor of PDF, actinonin, and confirmed our findings with a variety of other structurally different inhibitors. PDF has been shown to be a target of actinonin in bacteria (9), human cells (24), and plants (17).Here we show that inhibition of HsPDF function in mitochondria of human cell lines reduces mtDNA-encoded protein accumulation, new respiratory complex assembly, and energy production by the mitochondria. Aerobic glycolysis-dependent cell survival ensues upon disruption of HsPDF function. Therefore, HsPDF appears to fulfill a function in the mitochondria and to have a role in mtDNA-encoded protein-containing oxidative phosphorylation (OXPHOS) complex biogenesis.  相似文献   

5.
6.
The herpes simplex virus type 1 (HSV-1) gene UL12 encodes a conserved alkaline DNase with orthologues in all herpesviruses. The HSV-1 UL12 gene gives rise to two separately promoted 3′ coterminal mRNAs which encode distinct but related proteins: full-length UL12 and UL12.5, an amino-terminally truncated form that initiates at UL12 codon 127. Full-length UL12 localizes to the nucleus where it promotes the generation of mature viral genomes from larger precursors. In contrast, UL12.5 is predominantly mitochondrial and acts to trigger degradation of the mitochondrial genome early during infection. We examined the basis for these very different subcellular localization patterns. We confirmed an earlier report that the amino-terminal region of full-length UL12 is required for nuclear localization and provide evidence that multiple nuclear localization determinants are present in this region. In addition, we demonstrate that mitochondrial localization of UL12.5 relies largely on sequences located between UL12 residues 185 and 245 (UL12.5 residues 59 to 119). This region contains a sequence that resembles a typical mitochondrial matrix localization signal, and mutations that reduce the positive charge of this element severely impaired mitochondrial localization. Consistent with matrix localization, UL12.5 displayed a detergent extraction profile indistinguishable from that of the matrix protein cyclophilin D. Mitochondrial DNA depletion required the exonuclease activity of UL12.5, consistent with the idea that UL12.5 located within the matrix acts directly to destroy the mitochondrial genome. These results clarify how two highly related viral proteins are targeted to different subcellular locations with distinct functional consequences.All members of the Herpesviridae encode a conserved alkaline DNase that displays limited homology to bacteriophage λ red α (2, 24), an exonuclease that acts in conjunction with the synaptase red β to catalyze homologous recombination between DNA molecules (23). The most thoroughly characterized member of the herpesvirus alkaline nuclease family is encoded by the herpes simplex virus type 1 (HSV-1) gene UL12 (7, 9, 22). HSV-1 UL12 has both endo- and exonuclease activity (15-17, 36) and binds the viral single-stranded DNA binding protein ICP8 (37, 39) to form a recombinase that displays in vitro strand exchange activity similar to that for red α/β (27). UL12 localizes to the nucleus (26) where it plays an important, but as-of-yet ill-defined, role in promoting the production of mature packaged unit-length linear progeny viral DNA molecules (12, 20), perhaps via a recombination mechanism (27, 28). The importance of UL12 is documented by the observation that UL12 null mutants display a ca. 1,000-fold reduction in the production of infectious progeny virions (41).HSV-1 also produces an amino-terminally truncated UL12-related protein termed UL12.5, which is specified by a separately promoted mRNA that initiates within UL12 coding sequences (7, 9, 21). UL12.5 is translated in the same reading frame as UL12 but initiates at UL12 codon 127 and therefore lacks the first 126 amino acid residues of the full-length protein. UL12.5 retains the nuclease and ICP8 binding activities of UL12 (4, 14, 26) but does not accumulate to high levels in the nucleus (26) and is unable to efficiently substitute for UL12 in promoting viral genome maturation (14, 21). We recently showed that UL12.5 localizes predominantly to mitochondria, where it triggers massive degradation of the host mitochondrial genome early during HSV infection (31). Mammalian mitochondrial DNA (mt DNA) is a 16.5-kb double-stranded circle located within the mitochondrial matrix that encodes 13 proteins involved in oxidative phosphorylation and the RNA components of the mitochondrial translational apparatus (reviewed in reference 10). Inherited mutations that inactivate or deplete mt DNA impair oxidative phosphorylation, leading to a wide range of pathological conditions, including neuropathy and myopathy (reviewed in references 8 and 40). Thus, although the contribution of mt DNA depletion to the biology of HSV infection has yet to be determined, it likely has a major negative impact on host cell functions.UL12 and UL12.5 provide a striking example of a pair of highly related proteins that share a common biochemical activity yet differ markedly in subcellular location and biological function. The basis for their distinct subcellular localization patterns is of considerable interest, as the only difference in the primary sequences is that UL12.5 lacks the first 126 residues of UL12. Reuven et al. (26) demonstrated that this UL12-specific region contains one or more signals able to target enhanced green fluorescent protein (eGFP) to the nucleus. However, the determinants of the mitochondrial localization of UL12.5 have not been previously examined. Most proteins that are imported into the mitochondrial matrix bear a matrix targeting sequence that is located at or close to the amino terminus (reviewed in references 25 and 38). We speculated that UL12.5 bears such an amino-terminal matrix targeting sequence and that the function of this element is masked in the full-length UL12 protein by the UL12-specific amino-terminal extension, which contains the nuclear localization signal(s) (NLS). Our results broadly support this hypothesis and indicate that the mitochondrial localization sequence of UL12.5 is located ca. 60 residues from its N terminus.  相似文献   

7.
8.
Porin is the most abundant outer membrane (OM) protein of mitochondria. It forms the aqueous channel on the mitochondrial OM and mediates major metabolite flux between mitochondria and cytosol. Mitochondrial porin in Trypanosoma brucei, a unicellular parasitic protozoan and the causative agent of African trypanosomiasis, possesses a β-barrel structure similar to the bacterial OM porin OmpA. T. brucei porin (TbPorin) is present as a monomer as well as an oligomer on the mitochondrial OM, and its expression is developmentally regulated. In spite of its distinct structure, the TbPorin function is similar to those of other eukaryotic porins. TbPorin RNA interference (RNAi) reduced cell growth in both procyclic and bloodstream forms. The depletion of TbPorin decreased ATP production by inhibiting metabolite flux through the OM. Additionally, the level of trypanosome alternative oxidase (TAO) decreased, whereas the levels of cytochrome-dependent respiratory complexes III and IV increased in TbPorin-depleted mitochondria. Furthermore, the depletion of TbPorin reduced cellular respiration via TAO, which is not coupled with oxidative phosphorylation, but increased the capacity for cyanide-sensitive respiration. Together, these data reveal that TbPorin knockdown reduced the mitochondrial ATP level, which in turn increased the capacity of the cytochrome-dependent respiratory pathway (CP), in an attempt to compensate for the mitochondrial energy crisis. However, a simultaneous decrease in the substrate-level phosphorylation due to TbPorin RNAi caused growth inhibition in the procyclic form. We also found that the expressions of TAO and CP proteins are coordinately regulated in T. brucei according to mitochondrial energy demand.Trypanosoma brucei belongs to a group of parasitic protozoa that possess a single tubular mitochondrion with a concatenated structure of mitochondrial DNA known as kinetoplast (30). T. brucei is the infectious agent of the disease African trypanosomiasis, which is spread from one mammal to another by the bite of the tsetse fly (53). During transmission from the insect vector to the mammalian host and vice versa, the parasite undergoes various developmental stages accompanied by dramatic changes in mitochondrial activities (15). The bloodstream form that grows in mammalian blood uses glucose as its energy source and suppresses many mitochondrial activities. The bloodstream-form mitochondria lack cytochromes; thus, respiration in this form is solely dependent on the cytochrome-independent trypanosome alternative oxidase (TAO) (15). In contrast, the procyclic form that lives in the insect''s midgut possesses a developed mitochondrion with a full complement of the cytochrome-dependent respiratory system and a reduced level of TAO. The procyclic-form mitochondria produce ATP by both oxidative and substrate-level phosphorylations (7). On the other hand, the bloodstream-form mitochondria do not produce ATP but hydrolyze ATP to maintain the inner membrane (IM) potential (10, 33, 39, 48). Many of the mitochondrial IM- and matrix-localized proteins in T. brucei are well characterized (11, 29, 34, 43, 45). However, the mitochondrial outer membrane (OM) proteins in this group of parasitic protozoa have been poorly explored.Mitochondrial porin, which is also known as the voltage-dependent anion-selective channel (VDAC), is the most abundant protein in the OM (17, 28). The sizes and the secondary structures of this protein are very similar among different organisms. The VDAC possesses a N-terminal α-helical domain, and the rest of the protein consists of a number of amphiphilic β-strands, which form a barrel-like structure that integrates into the lipid bilayer (16, 17, 28). Recently, the three-dimensional structure of the human VDAC has been elucidated by nuclear magnetic resonance spectroscopy and X-ray crystallography, which showed a β-barrel architecture composed of 19 β-strands and the N-terminal α-helix located horizontally midway in the pore (5). Saccharomyces cerevisiae and Neurospora crassa VDACs also possess 16 to 19 β-strands, similar to the mammalian VDAC (17).The VDAC exists as different isomeric forms in different species (16, 19). In yeasts, there are two forms: VDAC1 and VDAC2. Only VDAC1 has the channel activity and is abundantly expressed (22, 23). Animals have three isoforms: VDAC1 to VDAC3. These isoforms showed more than 80% sequence homology among themselves. However, their expression levels and tissue specificities are different (16). Plants also have multiple isoforms of the VDAC with various expression levels under different pathological conditions (19). The VDAC plays a crucial role in regulated transport of ADP, ATP, Ca2+, and other metabolites in and out of mitochondria (17, 28, 41). Two ATP-binding sites found at the N- and C-terminal regions in the VDAC are critical for its function (54). Downregulation of VDAC expression disrupts mitochondrial energy production (22, 25). In contrast, overexpression of the VDAC in metazoa induces apoptosis, which can be blocked by compounds that inhibit its channel activity (1, 47).The OM of gram-negative bacteria also consists of various types of porins (24, 32, 40). Based on their structures and functions, they are divided into five groups. OmpA belongs to the small β-barrel integral membrane protein family, which is composed of eight β-strands. It is highly abundant and ubiquitous among most gram-negative bacteria (21). Other types of porins include general porin OmpF, which consists of 16 β-strands; substrate-specific porins, such as LamB or maltoporin, which contains 18 β-strands; receptor-type porin FhuA, the largest β-barrel, with 22 β-strands; and phospholipase A or OMPLA, an integral membrane enzyme containing 12 β-strands (21, 24, 32, 40). The OmpA plays important roles in bacterial conjugation, adhesion, invasion, and immune evasion and also acts as the receptor for several bacteriophages through its surface-exposed loops (44).Here, we show that the T. brucei mitochondrial porin (TbPorin) possesses a predicted β-barrel structure that has fewer β-strands than other mitochondrial porins but is similar to bacterial OmpA. TbPorin is crucial for mitochondrial energy production via both oxidative and substrate-level phosphorylations. The depletion of TbPorin reduced cell growth of the procyclic form as well as the bloodstream form. Furthermore, it reveals that depletion of mitochondrial ATP level by downregulation of porin alters the electron flow via TAO and the cytochrome-dependent pathway (CP) as well as the levels of proteins in these pathways.  相似文献   

9.
10.
The herpes simplex virus (HSV) virion host shutoff protein (vhs) encoded by gene UL41 is an mRNA-specific RNase that triggers accelerated degradation of host and viral mRNAs in infected cells. We report here that vhs is also able to modulate reporter gene expression without greatly altering the levels of the target mRNA in transient-transfection assays conducted in HeLa cells. We monitored the effects of vhs on a panel of bicistronic reporter constructs bearing a variety of internal ribosome entry sites (IRESs) located between two test cistrons. As expected, vhs inhibited the expression of the 5′ cistrons of all of these constructs; however, the response of the 3′ cistron varied with the IRES: expression driven from the wild-type EMCV IRES was strongly suppressed, while expression controlled by a mutant EMCV IRES and the cellular ApaF1, BiP, and DAP5 IRES elements was strongly activated. In addition, several HSV type 1 (HSV-1) 5′ untranslated region (5′ UTR) sequences also served as positive vhs response elements in this assay. IRES activation was also observed in 293 and HepG2 cells, but no such response was observed in Vero cells. Mutational analysis has yet to uncouple the ability of vhs to activate 3′ cistron expression from its shutoff activity. Remarkably, repression of 5′ cistron expression could be observed under conditions where the levels of the reporter RNA were not correspondingly reduced. These data provide strong evidence that vhs can modulate gene expression at the level of translation and that it is able to activate cap-independent translation through specific cis-acting elements.The virion host shutoff protein (vhs) encoded by herpes simplex virus (HSV) gene UL41 is an endoribonuclease that is packaged into the tegument of mature HSV virions. Once delivered into the cytoplasm of newly infected cells, vhs triggers shutoff of host protein synthesis, disruption of preexisting polysomes, and degradation of host mRNAs (reviewed in reference 62). The vhs-dependent shutoff system destabilizes many cellular and viral mRNAs (36, 46, 67). The rapid decline in host mRNA levels presumably helps viral mRNAs gain access to the cellular translational apparatus. In addition, the relatively short half-lives of viral mRNAs contribute to the sharp transitions between the successive phases of viral protein synthesis by tightly coupling changes in the rates of synthesis of viral mRNAs to altered mRNA levels (46). These effects enhance virus replication and may account for the modest reduction in virus yield displayed by vhs mutants in cultured Vero cells (55, 61).vhs also plays a critical role in HSV pathogenesis: vhs mutants are severely impaired for replication in the corneas and central nervous systems of mice and cannot efficiently establish or reactivate from latency (63, 65, 66). Mounting evidence indicates that this attenuation stems at least in part from an impaired ability to disarm elements of the innate and adaptive host immune responses (reviewed in reference 62). For example, vhs suppresses certain innate cellular antiviral responses, including production of proinflammatory cytokines and chemokines (68); dampens the type I interferon system (11, 45, 49, 78); and blocks activation of dendritic cells (58). Moreover, vhs mutants display enhanced virulence in knockout mice lacking type I interferon (IFN) receptors (37, 45) or Stat1 (48) and are hypersensitive to the antiviral effects of IFN in some cells in tissue culture (11, 49, 68). Thus, vhs is arguably a bona fide virulence factor.vhs present in extracts of HSV virions or purified from bacteria has nonspecific RNase activity capable of degrading all RNA substrates (15, 70, 71, 79). However, vhs is highly selective in vivo, targeting mRNAs and sparing other cytoplasmic RNAs (36, 46). In vivo and in mammalian whole-cell extracts, vhs-induced decay of at least some mRNAs initiates near regions of translation initiation and proceeds in an overall 5′-to-3′ direction (12, 13, 29, 52). Moreover, vhs binds to the translation initiation factors eIF4H, eIF4B, and eIF4A II, all components of the cap recognition factor eIF4F (10, 16, 17). Thus, it has been proposed that vhs selectively targets actively translated mRNAs through interactions with eIF4F components (17). Consistent with this hypothesis, recent data document that eIF4H is required for vhs activity in vivo (59).A previous report from this laboratory documented that the internal ribosome entry sites (IRESs) of the picornaviruses poliovirus and encephalomyocarditis virus (EMCV) strongly target vhs-induced RNA cleavage events to sequences immediately 3′ to the IRES in an in vitro translation system derived from rabbit reticulocyte lysates (RRL) (13). IRES elements are highly structured RNA sequences that are able to direct cap-independent translational initiation (reviewed in references 21, 25, 30, and 64). In the case of the poliovirus and EMCV elements, this is achieved by directly recruiting the eIF4F scaffolding protein eIF4G, thus bypassing the requirement for the cap-binding eIF4F subunit, eIF4E (reviewed in reference 30). Based on these data, we suggested that vhs is strongly targeted to the picornavirus IRES elements via interactions with eIF4 factors.A growing number of cellular mRNAs have been proposed to bear IRES elements in their 5′ untranslated regions (5′ UTRs). These include many that are involved in cellular stress responses, apoptosis, and cell cycle progression (24, 64, 74). Given the striking ability of picornavirus IRES elements to target vhs RNase activity in vitro, we asked whether viral and cellular IRES elements are able to modify the susceptibility of mRNAs to vhs in vivo. During the course of preliminary experiments designed to test this hypothesis, we unexpectedly discovered that vhs is able to strongly activate gene expression controlled by some cellular IRES elements and HSV 5′ UTR sequences in in vivo bicistronic reporter assays. These observations are the subject of the present report.  相似文献   

11.
12.
Soil substrate membrane systems allow for microcultivation of fastidious soil bacteria as mixed microbial communities. We isolated established microcolonies from these membranes by using fluorescence viability staining and micromanipulation. This approach facilitated the recovery of diverse, novel isolates, including the recalcitrant bacterium Leifsonia xyli, a plant pathogen that has never been isolated outside the host.The majority of bacterial species have never been recovered in the laboratory (1, 14, 19, 24). In the last decade, novel cultivation approaches have successfully been used to recover “unculturables” from a diverse range of divisions (23, 25, 29). Most strategies have targeted marine environments (4, 23, 25, 32), but soil offers the potential for the investigation of vast numbers of undescribed species (20, 29). Rapid advances have been made toward culturing soil bacteria by reformulating and diluting traditional media, extending incubation times, and using alternative gelling agents (8, 21, 29).The soil substrate membrane system (SSMS) is a diffusion chamber approach that uses extracts from the soil of interest as the growth substrate, thereby mimicking the environment under investigation (12). The SSMS enriches for slow-growing oligophiles, a proportion of which are subsequently capable of growing on complex media (23, 25, 27, 30, 32). However, the SSMS results in mixed microbial communities, with the consequent difficulty in isolation of individual microcolonies for further characterization (10).Micromanipulation has been widely used for the isolation of specific cell morphotypes for downstream applications in molecular diagnostics or proteomics (5, 15). This simple technology offers the opportunity to select established microcolonies of a specific morphotype from the SSMS when combined with fluorescence visualization (3, 11). Here, we have combined the SSMS, fluorescence viability staining, and advanced micromanipulation for targeted isolation of viable, microcolony-forming soil bacteria.  相似文献   

13.
14.
15.
Analysis of Lyme borreliosis (LB) spirochetes, using a novel multilocus sequence analysis scheme, revealed that OspA serotype 4 strains (a rodent-associated ecotype) of Borrelia garinii were sufficiently genetically distinct from bird-associated B. garinii strains to deserve species status. We suggest that OspA serotype 4 strains be raised to species status and named Borrelia bavariensis sp. nov. The rooted phylogenetic trees provide novel insights into the evolutionary history of LB spirochetes.Multilocus sequence typing (MLST) and multilocus sequence analysis (MLSA) have been shown to be powerful and pragmatic molecular methods for typing large numbers of microbial strains for population genetics studies, delineation of species, and assignment of strains to defined bacterial species (4, 13, 27, 40, 44). To date, MLST/MLSA schemes have been applied only to a few vector-borne microbial populations (1, 6, 30, 37, 40, 41, 47).Lyme borreliosis (LB) spirochetes comprise a diverse group of zoonotic bacteria which are transmitted among vertebrate hosts by ixodid (hard) ticks. The most common agents of human LB are Borrelia burgdorferi (sensu stricto), Borrelia afzelii, Borrelia garinii, Borrelia lusitaniae, and Borrelia spielmanii (7, 8, 12, 35). To date, 15 species have been named within the group of LB spirochetes (6, 31, 32, 37, 38, 41). While several of these LB species have been delineated using whole DNA-DNA hybridization (3, 20, 33), most ecological or epidemiological studies have been using single loci (5, 9-11, 29, 34, 36, 38, 42, 51, 53). Although some of these loci have been convenient for species assignment of strains or to address particular epidemiological questions, they may be unsuitable to resolve evolutionary relationships among LB species, because it is not possible to define any outgroup. For example, both the 5S-23S intergenic spacer (5S-23S IGS) and the gene encoding the outer surface protein A (ospA) are present only in LB spirochete genomes (36, 43). The advantage of using appropriate housekeeping genes of LB group spirochetes is that phylogenetic trees can be rooted with sequences of relapsing fever spirochetes. This renders the data amenable to detailed evolutionary studies of LB spirochetes.LB group spirochetes differ remarkably in their patterns and levels of host association, which are likely to affect their population structures (22, 24, 46, 48). Of the three main Eurasian Borrelia species, B. afzelii is adapted to rodents, whereas B. valaisiana and most strains of B. garinii are maintained by birds (12, 15, 16, 23, 26, 45). However, B. garinii OspA serotype 4 strains in Europe have been shown to be transmitted by rodents (17, 18) and, therefore, constitute a distinct ecotype within B. garinii. These strains have also been associated with high pathogenicity in humans, and their finer-scale geographical distribution seems highly focal (10, 34, 52, 53).In this study, we analyzed the intra- and interspecific phylogenetic relationships of B. burgdorferi, B. afzelii, B. garinii, B. valaisiana, B. lusitaniae, B. bissettii, and B. spielmanii by means of a novel MLSA scheme based on chromosomal housekeeping genes (30, 48).  相似文献   

16.
The ectopic overexpression of Bcl-2 restricts both influenza A virus-induced apoptosis and influenza A virus replication in MDCK cells, thus suggesting a role for Bcl-2 family members during infection. Here we report that influenza A virus cannot establish an apoptotic response without functional Bax, a downstream target of Bcl-2, and that both Bax and Bak are directly involved in influenza A virus replication and virus-induced cell death. Bak is substantially downregulated during influenza A virus infection in MDCK cells, and the knockout of Bak in mouse embryonic fibroblasts yields a dramatic rise in the rate of apoptotic death and a corresponding increase in levels of virus replication, suggesting that Bak suppresses both apoptosis and the replication of virus and that the virus suppresses Bak. Bax, however, is activated and translocates from the cytosol to the mitochondria; this activation is required for the efficient induction of apoptosis and virus replication. The knockout of Bax in mouse embryonic fibroblasts blocks the induction of apoptosis, restricts the infection-mediated activation of executioner caspases, and inhibits virus propagation. Bax knockout cells still die but by an alternative death pathway displaying characteristics of autophagy, similarly to our previous observation that influenza A virus infection in the presence of a pancaspase inhibitor leads to an increase in levels of autophagy. The knockout of Bax causes a retention of influenza A virus NP within the nucleus. We conclude that the cell and virus struggle to control apoptosis and autophagy, as appropriately timed apoptosis is important for the replication of influenza A virus.The pathology of influenza A virus infection usually arises from acute lymphopenia and inflammation of the lungs and airway columnar epithelial cells (23, 38). Influenza A virus induces apoptotic death in infected epithelial, lymphocyte, and phagocytic cells, and apoptosis is a source of tissue damage during infection (3, 22, 33) and increased susceptibility to bacterial pathogens postinfection (31). While the induction of apoptosis by influenza A virus has been well documented (4, 19-21, 28, 33, 37), the mechanisms of this interaction are not well understood. Two viral proteins, NS1 and PB1-F2, have been associated with viral killing of cells. NS1, originally characterized as being proapoptotic (34), was later identified as being an interferon antagonist, inhibiting the activation of several key antiviral responses and restricting the apoptotic response to infection (1, 10, 15, 18, 35, 39, 46). In contrast, PB1-F2 induces apoptosis primarily by localizing to the outer mitochondrial membrane, promoting cytochrome c release, and triggering the apoptotic cascade (43). This effect, however, is typically restricted to infected monocytes, leading to the hypothesis that PB1-F2 induces apoptosis specifically to clear the landscape of immune responders (5, 44). Although PB1-F2 activity does not directly manipulate virus replication or virus-induced apoptosis, PB1-F2 localization to the mitochondrial membrane during infection potentiates the apoptotic response in epithelial and fibroblastic cells through tBID signaling with proapoptotic Bcl-2 family protein members Bax and Bak (22, 43, 44).The Bcl-2 protein family consists of both pro- and antiapoptotic members that regulate cytochrome c release during mitochondrion-mediated apoptosis through the formation of pore-like channels in the outer mitochondrial membrane (12, 16). During the initiation of mitochondrion-mediated apoptosis, cytoplasmic Bid is cleaved to form tBID. This, in turn, activates proapoptotic Bax and Bak (40), which drive cytochrome c release and subsequent caspase activation. Bak is constitutively associated with the mitochondrial membrane, whereas inactive Bax is primarily cytosolic, translocating to the outer mitochondrial membrane only after activation (6). The activation of Bax and Bak results in homo- and heterodimer formation at the outer mitochondrial membrane, generating pores that facilitate mitochondrial membrane permeabilization and cytochrome c release (14, 17), leading to caspase activation and the apoptotic cascade (8). Antiapoptotic members of the Bcl-2 protein family, including Bcl-2, inhibit the activation of proapoptotic Bax and Bak primarily by sequestering inactive Bax and Bak monomers via interactions between their BH3 homology domains (7).Bcl-2 expression has been linked to decreased viral replication rates (26). Bcl-2 overexpression inhibits influenza A virus-induced cell death and reduces the titer and spread of newly formed virions (29). The activation of caspase-3 in the absence of sufficient Bcl-2 is critical to the influenza A virus life cycle. Both Bcl-2 expression and the lack of caspase activation during infection lead to the nuclear accumulation of influenza virus ribonucleoprotein (RNP) complexes, thereby leading to the improper assembly of progeny virions and a marked reduction in titers of infectious virus (26, 41, 42, 45).Here we show that influenza A virus induces mitochondrion-mediated (intrinsic-pathway) apoptosis signaled specifically through Bax and that this Bax signaling is essential for the maximum efficiency of virus propagation. In contrast, Bak expression is strongly downregulated during infection. Cells lacking Bak (while expressing Bax) display a much more severe apoptotic phenotype in response to infection and produce infectious virions at a higher rate than the wild type (WT), suggesting that Bak, which can suppress viral replication, is potentially downregulated by the virus. Our results indicate essential and opposing roles for Bax and Bak in both the response of cells to influenza A virus infection and the ability of the virus to maximize its own replicative potential.  相似文献   

17.
Human immunodeficiency virus type 1 (HIV-1) infects target cells by binding to CD4 and a chemokine receptor, most commonly CCR5. CXCR4 is a frequent alternative coreceptor (CoR) in subtype B and D HIV-1 infection, but the importance of many other alternative CoRs remains elusive. We have analyzed HIV-1 envelope (Env) proteins from 66 individuals infected with the major subtypes of HIV-1 to determine if virus entry into highly permissive NP-2 cell lines expressing most known alternative CoRs differed by HIV-1 subtype. We also performed linear regression analysis to determine if virus entry via the major CoR CCR5 correlated with use of any alternative CoR and if this correlation differed by subtype. Virus pseudotyped with subtype B Env showed robust entry via CCR3 that was highly correlated with CCR5 entry efficiency. By contrast, viruses pseudotyped with subtype A and C Env proteins were able to use the recently described alternative CoR FPRL1 more efficiently than CCR3, and use of FPRL1 was correlated with CCR5 entry. Subtype D Env was unable to use either CCR3 or FPRL1 efficiently, a unique pattern of alternative CoR use. These results suggest that each subtype of circulating HIV-1 may be subject to somewhat different selective pressures for Env-mediated entry into target cells and suggest that CCR3 may be used as a surrogate CoR by subtype B while FPRL1 may be used as a surrogate CoR by subtypes A and C. These data may provide insight into development of resistance to CCR5-targeted entry inhibitors and alternative entry pathways for each HIV-1 subtype.Human immunodeficiency virus type 1 (HIV-1) infects target cells by binding first to CD4 and then to a coreceptor (CoR), of which C-C chemokine receptor 5 (CCR5) is the most common (6, 53). CXCR4 is an additional CoR for up to 50% of subtype B and D HIV-1 isolates at very late stages of disease (4, 7, 28, 35). Many other seven-membrane-spanning G-protein-coupled receptors (GPCRs) have been identified as alternative CoRs when expressed on various target cell lines in vitro, including CCR1 (76, 79), CCR2b (24), CCR3 (3, 5, 17, 32, 60), CCR8 (18, 34, 38), GPR1 (27, 65), GPR15/BOB (22), CXCR5 (39), CXCR6/Bonzo/STRL33/TYMSTR (9, 22, 25, 45, 46), APJ (26), CMKLR1/ChemR23 (49, 62), FPLR1 (67, 68), RDC1 (66), and D6 (55). HIV-2 and simian immunodeficiency virus SIVmac isolates more frequently show expanded use of these alternative CoRs than HIV-1 isolates (12, 30, 51, 74), and evidence that alternative CoRs other than CXCR4 mediate infection of primary target cells by HIV-1 isolates is sparse (18, 30, 53, 81). Genetic deficiency in CCR5 expression is highly protective against HIV-1 transmission (21, 36), establishing CCR5 as the primary CoR. The importance of alternative CoRs other than CXCR4 has remained elusive despite many studies (1, 30, 70, 81). Expansion of CoR use from CCR5 to include CXCR4 is frequently associated with the ability to use additional alternative CoRs for viral entry (8, 16, 20, 63, 79) in most but not all studies (29, 33, 40, 77, 78). This finding suggests that the sequence changes in HIV-1 env required for use of CXCR4 as an additional or alternative CoR (14, 15, 31, 37, 41, 57) are likely to increase the potential to use other alternative CoRs.We have used the highly permissive NP-2/CD4 human glioma cell line developed by Soda et al. (69) to classify virus entry via the alternative CoRs CCR1, CCR3, CCR8, GPR1, CXCR6, APJ, CMKLR1/ChemR23, FPRL1, and CXCR4. Full-length molecular clones of 66 env genes from most prevalent HIV-1 subtypes were used to generate infectious virus pseudotypes expressing a luciferase reporter construct (19, 57). Two types of analysis were performed: the level of virus entry mediated by each alternative CoR and linear regression of entry mediated by CCR5 versus all other alternative CoRs. We thus were able to identify patterns of alternative CoR use that were subtype specific and to determine if use of any alternative CoR was correlated or independent of CCR5-mediated entry. The results obtained have implications for the evolution of env function, and the analyses revealed important differences between subtype B Env function and all other HIV-1 subtypes.  相似文献   

18.
19.
20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号