644 results on '"Luban, Jeremy"'
Search Results
202. Rescue of Multiple Viral Functions by a Second-Site Suppressor of a Human Immunodeficiency Virus Type 1 Nucleocapsid Mutation
- Author
-
Cimarelli, Andrea, primary, Sandin, Sara, additional, Höglund, Stefan, additional, and Luban, Jeremy, additional
- Published
- 2000
- Full Text
- View/download PDF
203. Basic Residues in Human Immunodeficiency Virus Type 1 Nucleocapsid Promote Virion Assembly via Interaction with RNA
- Author
-
Cimarelli, Andrea, primary, Sandin, Sara, additional, Höglund, Stefan, additional, and Luban, Jeremy, additional
- Published
- 2000
- Full Text
- View/download PDF
204. Human Immunodeficiency Virus Type 1 Gag Polyprotein Multimerization Requires the Nucleocapsid Domain and RNA and Is Promoted by the Capsid-Dimer Interface and the Basic Region of Matrix Protein
- Author
-
Burniston, Mark T., primary, Cimarelli, Andrea, additional, Colgan, John, additional, Curtis, Sean P., additional, and Luban, Jeremy, additional
- Published
- 1999
- Full Text
- View/download PDF
205. Translation Elongation Factor 1-Alpha Interacts Specifically with the Human Immunodeficiency Virus Type 1 Gag Polyprotein
- Author
-
Cimarelli, Andrea, primary and Luban, Jeremy, additional
- Published
- 1999
- Full Text
- View/download PDF
206. Human Immunodeficiency Virus Type 1 Replication Is Modulated by Host Cyclophilin A Expression Levels
- Author
-
Yin, Lei, primary, Braaten, Douglas, additional, and Luban, Jeremy, additional
- Published
- 1998
- Full Text
- View/download PDF
207. Sequence Note: The HIV Type 1 Replication Block in Nonhuman Primates Is Not Explained by Differences in Cyclophilin A Primary Structure
- Author
-
YIN, LEI, primary, BOUSSARD, SUZANNE, additional, ALLAN, JONATHAN, additional, and LUBAN, JEREMY, additional
- Published
- 1998
- Full Text
- View/download PDF
208. Inhibition of HIV-1 Replication by Cyclosporine A or Related Compounds Correlates with the Ability to Disrupt the Gag–Cyclophilin A Interaction
- Author
-
Franke, Ettaly Kara, primary and Luban, Jeremy, additional
- Published
- 1996
- Full Text
- View/download PDF
209. The yeast two-hybrid system for studying protein—protein interactions
- Author
-
Luban, Jeremy, primary and Goff, Stephen P, additional
- Published
- 1995
- Full Text
- View/download PDF
210. Evidence for biphasic uncoating during HIV-1 infection from a novel imaging assay.
- Author
-
Hongzhan Xu, Franks, Tamera, Gibson, Gregory, Huber, Kelly, Rahm, Nadia, De Castillia, Caterina Strambio, Luban, Jeremy, Aiken, Christopher, Watkins, Simon, Sluis-Cremer, Nicolas, and Ambrose, Zandrea
- Subjects
VIRAL proteins ,VIRION ,MICROSCOPY ,HIV infections ,RNA - Abstract
Background: Uncoating of the HIV-1 core plays a critical role during early post-fusion stages of infection but is poorly understood. Microscopy-based assays are unable to easily distinguish between intact and partially uncoated viral cores. Results: In this study, we used 5-ethynyl uridine (EU) to label viral-associated RNA during HIV production. At early time points after infection with EU-labeled virions, the viral-associated RNA was stained with an EU-specific dye and was detected by confocal microscopy together with viral proteins. We observed that detection of the viral-associated RNA was specific for EU-labeled virions, was detected only after viral fusion with target cells, and occurred after an initial opening of the core. In vitro staining of cores showed that the opening of the core allowed the small molecule dye, but not RNase A or antibodies, inside. Also, staining of the viral-associated RNA, which is co-localized with nucleocapsid, decays over time after viral infection. The decay rate of RNA staining is dependent on capsid (CA) stability, which was altered by CA mutations or a small molecule inducer of HIV-1 uncoating. While the staining of EU-labeled RNA was not affected by inhibition of reverse transcription, the kinetics of core opening of different CA mutants correlated with initiation of reverse transcription. Analysis of the E45A CA mutant suggests that initial core opening is independent of complete capsid disassembly. Conclusions: Taken together, our results establish a novel RNA accessibility-based assay that detects an early event in HIV-1 uncoating and can be used to further define this process. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
211. TRIM5 structure, HIV-1 capsid recognition, and innate immune signaling.
- Author
-
Grütter, Markus G and Luban, Jeremy
- Subjects
HIV ,CAPSIDS ,NATURAL immunity ,RETROVIRUS diseases ,VIRION ,VIRAL genomes ,UBIQUITIN ligases ,CHEMOKINES - Abstract
TRIM5 is a restriction factor that blocks retrovirus infection soon after the virion core enters the cell cytoplasm. Restriction activity is targeted to the virion core via recognition of the capsid protein lattice that encases the viral genomic RNA. In common with all of the many TRIM family members, TRIM5 has RING, B-box, and coiled-coil domains. As an E3 ubiquitin ligase TRIM5 cooperates with the heterodimeric E2, UBC13/UEV1A, to activate the TAK1 (MAP3K7) kinase, NF-κB and AP-1 signaling, and the transcription of inflammatory cytokines and chemokines. TAK1, UBC13, and UEV1A all contribute to TRIM5-mediated retrovirus restriction activity. Interaction of the carboxy-terminal PRYSPRY or cyclophilin domains of TRIM5 with the retroviral capsid lattice stimulates the formation of a complementary lattice by TRIM5, with greatly increased TRIM5 E3 activity, and host cell signal transduction. Structural and biochemical studies on TRIM5 have opened a much needed window on how the innate immune system detects the distinct molecular features of HIV-1 and other retroviruses. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
212. HERV-H RNA is abundant in human embryonic stem cells and a precise marker for pluripotency.
- Author
-
Santoni, Federico A., Guerra, Jessica, and Luban, Jeremy
- Subjects
EMBRYONIC stem cells ,TRANSCRIPTION factors ,RETROVIRUSES ,CHROMATIN ,GENOMES - Abstract
Background: Certain post-translational modifications to histones, including H3K4me3, as well as binding sites for the transcription factor STAT1, predict the site of integration of exogenous gamma-retroviruses with great accuracy and cell-type specificity. Statistical methods that were used to identify chromatin features that predict exogenous gamma-retrovirus integration site selection were exploited here to determine whether cell type-specific chromatin markers are enriched in the vicinity of endogenous retroviruses (ERVs). Results: Among retro-elements in the human genome, the gamma-retrovirus HERV-H was highly associated with H3K4me3, though this association was only observed in embryonic stem (ES) cells (p < 10
-300 ) and, to a lesser extent, in induced pluripotent stem (iPS) cells. No significant association was observed in nearly 40 differentiated cell types, nor was any association observed with other retro-elements. Similar strong association was observed between HERV-H and the binding sites within ES cells for the pluripotency transcription factors NANOG, OCT4, and SOX2. NANOG binding sites were located within the HERV-H 5′LTR itself. OCT4 and SOX2 binding sites were within 1 kB and 2 kB of the 5′LTR, respectively. In keeping with these observations, HERV-H RNA constituted 2% of all poly A RNA in ES cells. As ES cells progressed down a differentiation pathway, the levels of HERV-H RNA decreased progressively. RNA-Seq datasets showed HERV-H transcripts to be over 5 kB in length and to have the structure 5′LTR-gag-pro-3′LTR, with no evidence of splicing and no intact open reading frames. Conclusion: The developmental regulation of HERV-H expression, the association of HERV-H with binding sites for pluripotency transcription factors, and the extremely high levels of HERV-H RNA in human ES cells suggest that HERV-H contributes to pluripotency in human cells. Proximity of HERV-H to binding sites for pluripotency transcription factors within ES cells might be due to retention of the same chromatin features that determined the site of integration of the ancestral, exogenous, gamma-retrovirus that gave rise to HERV-H in the distant past. Retention of these markers, or, alternatively, recruitment of them to the site of the established provirus, may have acted post-integration to fix the provirus within the germ-line of the host species. Either way, HERV-H RNA provides a specific marker for pluripotency in human cells. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
213. Nef Decreases HIV-1 Sensitivity to Neutralizing Antibodies that Target the Membrane-proximal External Region of TMgp41.
- Author
-
J. Lai, Rachel P., Jin Yan, Heeney, Jonathan, McClure, Myra O., Göttlinger, Heinrich, Luban, Jeremy, and Pizzato, Massimo
- Abstract
Primate lentivirus nef is required for sustained virus replication in vivo and accelerated progression to AIDS. While exploring the mechanism by which Nef increases the infectivity of cell-free virions, we investigated a functional link between Nef and Env. Since we failed to detect an effect of Nef on the quantity of virion-associated Env, we searched for qualitative changes by examining whether Nef alters HIV-1 sensitivity to agents that target distinct features of Env. Nef conferred as much as 50- fold resistance to 2F5 and 4E10, two potent neutralizing monoclonal antibodies (nAbs) that target the membrane proximal external region (MPER) of TMgp41. In contrast, Nef had no effect on HIV-1 neutralization by MPER-specific nAb Z13e1, by the peptide inhibitor T20, nor by a panel of nAbs and other reagents targeting gp120. Resistance to neutralization by 2F5 and 4E10 was observed with Nef from a diverse range of HIV-1 and SIV isolates, as well as with HIV-1 virions bearing Env from CCR5- and CXCR4-tropic viruses, clade B and C viruses, or primary isolates. Functional analysis of a panel of Nef mutants revealed that this activity requires Nef myristoylation but that it is genetically separable from other Nef functions such as the ability to enhance virus infectivity and to downregulate CD4. Glycosylated-Gag from MoMLV substituted for Nef in conferring resistance to 2F5 and 4E10, indicating that this activity is conserved in a retrovirus that does not encode Nef. Given the reported membrane-dependence of MPER-recognition by 2F5 and 4E10, in contrast to the membrane-independence of Z13e1, the data here is consistent with a model in which Nef alters MPER recognition in the context of the virion membrane. Indeed, Nef and Glycosylated-Gag decreased the efficiency of virion capture by 2F5 and 4E10, but not by other nAbs. These studies demonstrate that Nef protects lentiviruses from one of the most broadly-acting classes of neutralizing antibodies. This newly discovered activity for Nef has important implications for anti-HIV-1 immunity and AIDS pathogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
214. TRIM5 is an innate immune sensor for the retrovirus capsid lattice.
- Author
-
Pertel, Thomas, Hausmann, Stéphane, Morger, Damien, Züger, Sara, Guerra, Jessica, Lascano, Josefina, Reinhard, Christian, Santoni, Federico A., Uchil, Pradeep D., Chatel, Laurence, Bisiaux, Aurélie, Albert, Matthew L., Strambio-De-Castillia, Caterina, Mothes, Walther, Pizzato, Massimo, Grütter, Markus G., and Luban, Jeremy
- Subjects
HIV ,RETROVIRUSES ,CYTOPLASM ,ENDOTOXINS ,ENZYMES ,UBIQUITIN - Abstract
TRIM5 is a RING domain-E3 ubiquitin ligase that restricts infection by human immunodeficiency virus (HIV)-1 and other retroviruses immediately following virus invasion of the target cell cytoplasm. Antiviral potency correlates with TRIM5 avidity for the retrovirion capsid lattice and several reports indicate that TRIM5 has a role in signal transduction, but the precise mechanism of restriction is unknown. Here we demonstrate that TRIM5 promotes innate immune signalling and that this activity is amplified by retroviral infection and interaction with the capsid lattice. Acting with the heterodimeric, ubiquitin-conjugating enzyme UBC13-UEV1A (also known as UBE2N-UBE2V1), TRIM5 catalyses the synthesis of unattached K63-linked ubiquitin chains that activate the TAK1 (also known as MAP3K7) kinase complex and stimulate AP-1 and NFκB signalling. Interaction with the HIV-1 capsid lattice greatly enhances the UBC13-UEV1A-dependent E3 activity of TRIM5 and challenge with retroviruses induces the transcription of AP-1 and NF-κB-dependent factors with a magnitude that tracks with TRIM5 avidity for the invading capsid. Finally, TAK1 and UBC13-UEV1A contribute to capsid-specific restriction by TRIM5. Thus, the retroviral restriction factor TRIM5 has two additional activities that are linked to restriction: it constitutively promotes innate immune signalling and it acts as a pattern recognition receptor specific for the retrovirus capsid lattice. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
215. Efficient IgM assembly and secretion require the plasma cell induced endoplasmic reticulum protein pERp1.
- Author
-
Van Anken, Eelco, Pena, Florentina, Hafkemeijer, Nicole, Christis, Chantal, Romijn, Edwin P., Grauschopf, Ulla, Oorschot, Viola M. J., Pertel, Thomas, Engels, Sander, An Ora, Lástun, Viorica, Glockshuber, Rudi, Klumperman, Judith, Heck, Albert J. R., Luban, Jeremy, and Braakman, Ineke
- Subjects
PLASMA cells ,IMMUNOGLOBULINS ,ENDOPLASMIC reticulum ,OXIDOREDUCTASES ,LYMPHOCYTE transformation ,CYSTATHIONINE gamma-lyase - Abstract
Plasma cells daily secrete their own mass in antibodies, which fold and assemble in the endoplasmic reticulum (ER). To reach these levels, cells require pERp1, a novel lymphocyte-specific small ER-resident protein, which attains expression levels as high as BiP when B cells differentiate into plasma cells. Although pERp1 has no homology with known ER proteins, it does contain a CXXC motif typical for oxidoreductases. In steady state, the CXXC cysteines are locked by two parallel disulfide bonds with a downstream C(X)
6 C motif, and pERpi displays only modest oxidoreductase activity. pERp1 emerged as a dedicated folding factor for IgM, associating with both heavy and light chains and promoting assembly and secretion of mature IgM. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
216. Lentiviral Vector Gene Transfer Is Limited by the Proteasome at Postentry Steps in Various Types of Stem Cells.
- Author
-
de Sio, Francesca Romana Santoni, Gritti, Angela, Cascio, Paolo, Neri, Margherita, Sampaolesi, Maurilio, Galli, Cesare, Luban, Jeremy, and Naldini, Luigi
- Subjects
GENETIC transformation ,STEM cells ,LENTIVIRUSES ,VIRUS diseases ,GENE therapy ,CELLULAR therapy ,TRANSPLANTATION of organs, tissues, etc. - Abstract
The isolation of human embryonic and somatic stem cells of different types has made it possible to design novel gene and cell replacement therapies. Vectors derived from retro/lentiviruses are used to stably introduce genes into stem cells and their progeny. However, the permissivity to retroviral infection varies among cell types. We previously showed that hematopoietic stem cells are poorly permissive to human immunodeficiency virus (HIV)-derived vectors and that pharmacological inhibition of the proteasome strongly enhances gene transfer. Here we report that the proteasome limits lentiviral gene transfer in all stem cell types tested, including embryonic, mesenchymal, and neural, of both human and mouse origin. Remarkably, this inhibitory activity was sharply reduced upon differentiation of the stem cells, suggesting that it represents a novel feature of the stem cell/immature progenitor phenotype. Proteasome-mediated inhibition was specific for lentiviral vectors and occurred at a postentry infection step. It was not mediated by activation of nuclear factor-κB, a major signaling pathway modulated by the proteasome, and did not correlate with high proteasome activity. Interaction of the virion core with cyclophilin A was required to maximize the effect of proteasome inhibitor on the infection pathway. These findings are relevant to uncover new mediators of HIV gene transfer and help in designing more effective protocols for the genetic modification of stem cells. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
217. The retroviral restriction factor TRIM5α.
- Author
-
Sebastian, Sarah and Luban, Jeremy
- Abstract
Retroviruses are obligate intracellular parasites that have coevolved with their hosts for millions of years. It is therefore not surprising that retroviruses take advantage of numerous host factors during their life cycle. In addition to positive cellular factors that are of use to the virus, host cells have also evolved intracellular proteins to antagonize the retroviral replication cycle. Such inhibitory cellular factors have been called retroviral restriction factors. Recently, several such restriction factors have been cloned, including Friend virus susceptibility factor 1, apolipoprotein B mRNA-editing enzyme catalytic proteins 3F and 3G, and ZAP. Here, we review the explosion of publications from the past 2 years concerning TRIM5, a host factor that potently inhibits HIV-1 and other retroviruses. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
218. Cyclophilin, TRIM5, and innate immunity to HIV-1
- Author
-
Sokolskaja, Elena and Luban, Jeremy
- Subjects
- *
CYCLOPHILINS , *CIS-trans-isomerases , *HIV , *HIV infections , *IMMUNITY - Abstract
The peptidyl-prolyl isomerase cyclophilin A (CypA) binds a proline-rich loop on the surface of HIV-1 capsid (CA). This interaction increases HIV-1 infectivity in humans but promotes an anti-HIV-1 restriction activity in non-human primates. Efforts to understand these paradoxical effects of cyclophilin, along with more targeted approaches to uncover the genetic basis for HIV-1 restriction, led to the discovery of TRIM5 (tripartite motif protein 5), a CA-specific receptor for the retroviral core. The ensuing TRIM5 publication flurry established a paradigm of innate immunity in which the protein lattice of an invading retroviral core, rather than double-stranded RNA or lipopolysaccharide, is recognized by a multimeric, cytoplasmic receptor. CypA modulates HIV-1 virion core detection by this class of innate pattern recognition molecule, apparently by inducing subtle shifts in CA conformation. [Copyright &y& Elsevier]
- Published
- 2006
- Full Text
- View/download PDF
219. Cyclophilin A is required for TRIM5α-mediated resistance to HIV-1 in Old World monkey cells.
- Author
-
Berthoux, Lionel, Sebastian, Sarah, Sokolskaja, Elena, and Luban, Jeremy
- Subjects
CYCLOPHILINS ,LABORATORY monkeys ,CELLS ,HIV infections ,NUCLEIC acids ,RNA ,RHESUS monkeys - Abstract
The peptidyl-prolyl isomerase cyclophilin A (CypA) embraces an exposed, proline-rich loop on HIV-1 capsid (CA) and renders reverse transcription complexes resistant to an antiviral activity in human cells. A CypA fusion with TRIM5 that is unique to New World owl monkeys also targets HIV-1 CA, but this interaction potently inhibits infection. A similar block to HIV-1 infection in Old World monkeys is attributable to the a isoform of the TRIM5 orthologue in these species. To determine whether HIV-1 restriction by Old World monkey TRIM5α is modulated by the CA-CypA interaction, RNA interference was used to disrupt CypA in cells from African green monkeys and rhesus macaques. HIV-1 infectivity increased in response to CypA knock-down to the same extent that it increased in response to TRIMS knock-down. CypA knock-down eliminated the HIV-1 stimulatory effect of cyclosporin A (CsA), a competitive inhibitor of the CypA-CA interaction, or of CA mutants that block binding to CypA but caused no change in titer of retroviruses that don't interact with CypA. Simultaneous knock-down of both CypA and TRIMS caused minimal additional increase in titer, suggesting that CypA inhibits HIV-1 replication in these cells because it is required for CA recognition by TRIMSa. Finally, CsA increased HIV-1 titer in otherwise nonrestrictive feline cells but only after these cells were transduced with Old World monkey TRIM5α. Thus, CypA is required for HIV-1 restriction by Old World monkey orthologues of TRIM5α. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
220. Selection for Loss of Ref1Activity in Human Cells Releases Human Immunodeficiency Virus Type 1 from Cyclophilin A Dependence during Infection.
- Author
-
Sayah, David M. and Luban, Jeremy
- Subjects
- *
HIV , *HTLV , *CYCLOPHILINS , *VIROLOGY , *MICROBIOLOGY , *MICROORGANISMS - Abstract
Capsid (CA)-specific restrictions are determinants of retroviral tropism in mammalian cells. One such restriction, human Ref1, targets strains of murine leukemia virus bearing an arginine at CA residue 110 (N-MLV), resulting in decreased accumulation of viral cDNA. The cellular factors accounting for Reft activity are unknown. As2O3 increases N-MLV titer in Ref1-positive cells, possibly by counteracting Ref1. Restriction factor saturation experiments suggest that Reft may also target human immunodeficiency virus type 1 (HIV-1), but only if its CA is not bound to the cellular protein cyclophilin A (CypA). As a step towards understanding the genetic determinants of Ref1, we subjected Ref1-positive TE671 cells to three sequential rounds of selection with N-MLV reporter viruses. We isolated a subclone, 17H1, that was permissive for N-MLV infection and therefore deficient in Reft activity. Stimulation of N-MLV replication by As2O3 was attenuated in 17H1, confirming that the drug acts by overcoming Ref1 activity. HIV-1 infection of 17H1 cells was resistant to disruption of the CA-CypA interaction, demonstrating that Ref1 restricts CypA-free HIV-1. Our results suggest that interaction with CypA evolved to protect HIV-1 from this human antiviral activity. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
221. Cyclophilin A retrotransposition into TRIM5 explains owl monkey resistance to HIV-1.
- Author
-
Sayah, David M., Sokolskaja, Elena, Berthoux, Lionel, and Luban, Jeremy
- Subjects
CYCLOPHILINS ,THREE-striped night monkey ,HIV ,PRIMATES ,NATURAL immunity ,MESSENGER RNA - Abstract
In Old World primates, TRIM5-a confers a potent block to human immunodeficiency virus type 1 (HIV-1) infection that acts after virus entry into cells. Cyclophilin A (CypA) binding to viral capsid protects HIV-1 from a similar activity in human cells. Among New World primates, only owl monkeys exhibit post-entry restriction of HIV-1 (ref. 1). Paradoxically, the barrier to HIV-1 in owl monkey cells is released by capsid mutants or drugs that disrupt capsid interaction with CypA. Here we show that knockdown of owl monkey CypA by RNA interference (RNAi) correlates with suppression of anti-HIV-1 activity. However, reintroduction of CypA protein to RNAi-treated cells did not restore antiviral activity. A search for additional RNAi targets unearthed TRIMCyp, an RNAi-responsive messenger RNA encoding a TRIM5-CypA fusion protein. TRIMCyp accounts for post-entry restriction of HIV-1 in owl monkeys and blocks HIV-1 infection when transferred to otherwise infectable human or rat cells. It seems that TRIMCyp arose after the divergence of New and Old World primates when a LINE-1 retrotransposon catalysed the insertion of a CypA complementary DNA into the TRIM5 locus. This is the first vertebrate example of a chimaeric gene generated by this mechanism of exon shuffling. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
222. AIF and cyclophilin A cooperate in apoptosis-associated chromatinolysis.
- Author
-
Candé, Céline, Vahsen, Nicola, Kouranti, Ilektra, Schmitt, Elise, Daugas, Eric, Spahr, Chris, Luban, Jeremy, Kroemer, Romano T., Giordanetto, Fabrizio, Garrido, Carmen, Penninger, Josef M., and Kroemer, Guido
- Subjects
MITOCHONDRIA ,CYCLOPHILINS ,MASS spectrometry ,APOPTOSIS ,DNA damage - Abstract
Cyclophilin A (CypA) was determined to interact with apoptosis-inducing factor (AIF) by mass spectroscopy, coimmunoprecipitation, pull-down assays, and molecular modeling. During the initial, caspase-independent stage of chromatin condensation that accompanies apoptosis, AIF and CypA were found to coimmunolocalize in the nucleus. Recombinant AIF and CypA proteins synergized in vitro in the degradation of plasmid DNA, as well as in the capacity to induce DNA loss in purified nuclei. The apoptogenic cooperation between AIF and CypA did not rely on the CypA peptidyl-prolyl cis-trans isomerase activity. In Cyp-expressing cells, AIF overexpression augmented apoptotic chromatinolysis. The AIF-dependent large-scale DNA fragmentation was less pronounced in CypA knockout cells as compared to controls. AIF mutants lacking the CypA-binding domain were inefficient apoptosis sensitizers in transfection experiments. Moreover, AIF failed to sensitize CypA knockout cells to apoptosis induction, and this defect in the AIF response was reversed by reintroduction of the CypA gene into CypA-deficient cells. In summary, AIF and CypA collaborate in chromatinolysis.Oncogene (2004) 23, 1514-1521. doi:10.1038/sj.onc.1207279 Published online 12 January 2004 [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
223. Cyclophilin A modulates the sensitivity of HIV-1 to host restriction factors.
- Author
-
Towers, Greg J., Hatziioannou, Theodora, Cowan, Simone, Goff, Stephen P., Luban, Jeremy, and Bieniasz, Paul D.
- Subjects
CYCLOPHILINS ,HIV - Abstract
Many mammalian species express restriction factors that confer host resistance to retroviral infection. Here we show that HIV-1 sensitivity to restriction factors is modulated by cyclophilin A (CypA), a host cell protein that binds the HIV-1 capsid protein (CA). In certain nonhuman primate cells, the CA-CypA interaction is essential for restriction: HIV-1 infectivity is increased >100-fold by cyclosporin A (CsA), a competitive inhibitor of the interaction, or by an HIV-1 CA mutation that disrupts CypA binding. Conversely, disruption of CA-CypA interaction in human cells reveals that CypA protects HIV-1 from the Ref-1 restriction factor. These findings suggest that HIV-1 has co-opted a host cell protein to counteract restriction factors expressed by human cells and that this adaptation can confer sensitivity to restriction in unnatural hosts. Manipulation of HIV-1 CA recognition by restriction factors promises to advance animal models and new therapeutic strategies for HIV-1 and AIDS. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
224. Cyclophilin A regulates HIV-1 infectivity, as demonstrated by gene targeting in human T cells.
- Author
-
Braaten, Douglas and Luban, Jeremy
- Subjects
- *
CYCLOPHILINS , *HIV , *RETROVIRUSES , *ISOMERASES , *T cells , *LYMPHOCYTES - Abstract
The human immunodeficiency virus type 1 (HIV-1) Gag polyprotein binds most members of the cyclophilin family of peptidyl-prolyl isomerases. Of 15 known human cyclophilins, cyclophilin A (CypA) has been the focus of investigation because it was detected in HIV-1 virions. To determine whether CypA promotes HIV-1 replication, we deleted the gene encoding CypA (PPIA) in human CD4+ T cells by homologous recombination. HIV-1 replication in PPIA-/- cells was decreased and not inhibited further by cyclosporin or gag mutations that disrupt Gag's interaction with cyclophilins, indicating that no other cyclophilin family members promote HIV-1 replication. The defective replication phenotype was specific for wild-type HIV-1 since HIV-2/SIV isolates, as well as HIV-1 bearing a gag mutation that confers cyclosporin resistance, replicated the same in PPIA+/+ and PPIA-/- cells. Stable re-expression of CypA in PPIA-/- cells restored HIV-1 replication to an extent that correlated with steady-state levels of CypA. Finally, virions from PPIA-/- cells possessed no obvious biochemical abnormalities but were less infectious than virions from wild-type cells. These data formally demonstrate that CypA regulates the infectivity of HIV-1 virions. [ABSTRACT FROM AUTHOR]
- Published
- 2001
- Full Text
- View/download PDF
225. Targeting of the Tec Kinase ITK Drives Resolution of T Cell–Mediated Colitis and Emerges as Potential Therapeutic Option in Ulcerative Colitis.
- Author
-
Lechner, Kristina, Mott, Stefanie, Al-Saifi, Ragheed, Knipfer, Lisa, Wirtz, Stefan, Atreya, Raja, Vieth, Michael, Rath, Timo, Fraass, Tina, Winter, Zoltan, August, Avery, Luban, Jeremy, Zimmermann, Valérie S., Weigmann, Benno, and Neurath, Markus F.
- Abstract
The molecular checkpoints driving T cell activation and cytokine responses in ulcerative colitis (UC) are incompletely understood. Here, we studied the Tec kinase ITK in UC. We analyzed patients with inflammatory bowel disease (n = 223) and evaluated ITK activity as well as the functional effects of cyclosporine-A (CsA). In addition, 3 independent murine colitis models were used to investigate the functional role of ITK. Finally, the activity of ITK was blocked via pharmacological inhibitors and genetically engineered mice. Readout parameters were mini-endoscopy, histopathology, mucosal T cell apoptosis, and cytokine production. We found an expansion of pITK-expressing mucosal CD4
+ T cells in UC rather than Crohn's disease that correlated with disease severity. CsA suppressed activation of ITK in cultured CD4+ T cells and calcineurin-containing microclusters adjacent to the T cell receptor signaling complex. Functionally, the capacity of CsA to suppress activity of experimental colitis was critically dependent on ITK. Genetic inactivation of Itk via gene targeting or induction of allele-sensitive Itk mutants prevented experimental colitis in 3 colitis models, and treatment with pharmacological ITK blockers suppressed established colitis. In addition, ITK controlled apoptosis and activation of mucosal Th2 and Th17 lymphocytes via NFATc2 signaling pathways. ITK activation was detected in UC and could be down-regulated in cultured T cells by CsA administration. Selective targeting of ITK emerges as an attractive approach for treatment of chronic intestinal inflammation and potentially UC by driving resolution of mucosal inflammation. [Display omitted] ITK is highly activated in patients with ulcerative colitis only and cyclosporine-A suppresses ITK activity. Therapeutic modulation of the cyclosporine-A target ITK might be helpful for optimized therapy in ulcerative colitis, as inactivation of ITK leads to resolution of intestinal mucosal inflammation. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
226. Cyclophilin A facilitates HIV-1 integration.
- Author
-
Padron, Adrian, Dwivedi, Richa, Chakraborty, Rajasree, Prakash, Prem, Kyusik Kim, Jiong Shi, Jinwoo Ahn, Jui Pandhare, Luban, Jeremy, Aiken, Christopher, Balasubramaniam, Muthukumar, and Dash, Chandravanu
- Abstract
Cyclophilin A (CypA) binds to the HIV-1 capsid to facilitate reverse transcription and nuclear entry and counter the antiviral activity of TRIM5a. Interestingly, recent studies suggest that the capsid enters the nucleus of an infected cell and uncoats prior to integration. We have previously reported that the capsid protein regulates HIV-1 integration. Therefore, we probed whether CypA-capsid interaction also regulates this post-nuclear entry step. First, we challenged CypA-expressing (CypA+/+) and CypA-depleted (CypA-/-) cells with HIV-1 and quantified the levels of provirus. CypA-depletion significantly reduced integration, an effect that was independent of CypA's effect on reverse transcription, nuclear entry, and the presence or absence of TRIM5a. In addition, cyclosporin A, an inhibitor that disrupts CypA-capsid binding, inhibited proviral integration in CypA+/+ cells but not in CypA-/- cells. HIV-1 capsid mutants (G89V and P90A) deficient in CypA binding were also blocked at the integration step in CypA+/+ cells but not in CypA-/- cells. Then, to understand the mechanism, we assessed the integration activity of the HIV-1 preintegration complexes (PICs) extracted from acutely infected cells. PICs from CypA-/- cells retained lower integration activity in vitro compared to those from CypA+/+ cells. PICs from cells depleted of both CypA and TRIM5a also had lower activity, suggesting that CypA's effect on PIC was independent of TRIM5a. Finally, CypA protein specifically stimulated PIC activity, as this effect was significantly blocked by CsA. Collectively, these results provide strong evidence that CypA directly promotes HIV-1 integration, a previously unknown role of this host factor in the nucleus of an infected cell. IMPORTANCE Interaction between the HIV-1 capsid and host cellular factors is essential for infection. However, the molecular details and functional consequences of viral-host factor interactions during HIV-1 infection are not fully understood. Over 30 years ago, Cyclophilin A (CypA) was identified as the first host protein to bind to the HIV-1 capsid. Now it is established that CypA-capsid interaction promotes reverse transcription and nuclear entry of HIV-1. In addition, CypA blocks TRIM5a-mediated restriction of HIV-1. In this report, we show that CypA promotes the post-nuclear entry step of HIV-1 integration by binding to the viral capsid. Notably, we show that CypA stimulates the viral DNA integration activity of the HIV-1 preintegration complex. Collectively, our studies identify a novel role of CypA during the early steps of HIV-1 infection. This new knowledge is important because recent reports suggest that an operationally intact HIV-1 capsid enters the nucleus of an infected cell. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
227. Influence of Different Glycoproteins and of the Virion Core on SERINC5 Antiviral Activity.
- Author
-
Diehl, William E., Guney, Mehmet H., Vanzo, Teresa, Kyawe, Pyae P., White, Judith M., Pizzato, Massimo, and Luban, Jeremy
- Subjects
RETROVIRUSES ,ARENAVIRUSES ,GLYCOPROTEINS ,EQUINE infectious anemia ,MOUSE leukemia viruses ,SIMIAN viruses ,VESICULAR stomatitis ,VIRION - Abstract
Host plasma membrane protein SERINC5 is incorporated into budding retrovirus particles where it blocks subsequent entry into susceptible target cells. Three structurally unrelated proteins encoded by diverse retroviruses, human immunodeficiency virus type 1 (HIV-1) Nef, equine infectious anemia virus (EIAV) S2, and ecotropic murine leukemia virus (MLV) GlycoGag, disrupt SERINC5 antiviral activity by redirecting SERINC5 from the site of virion assembly on the plasma membrane to an internal RAB7+ endosomal compartment. Pseudotyping retroviruses with particular glycoproteins, e.g., vesicular stomatitis virus glycoprotein (VSV G), renders the infectivity of particles resistant to inhibition by virion-associated SERINC5. To better understand viral determinants for SERINC5-sensitivity, the effect of SERINC5 was assessed using HIV-1, MLV, and Mason-Pfizer monkey virus (M-PMV) virion cores, pseudotyped with glycoproteins from Arenavirus, Coronavirus, Filovirus, Rhabdovirus, Paramyxovirus, and Orthomyxovirus genera. SERINC5 restricted virions pseudotyped with glycoproteins from several retroviruses, an orthomyxovirus, a rhabdovirus, a paramyxovirus, and an arenavirus. Infectivity of particles pseudotyped with HIV-1, amphotropic-MLV (A-MLV), or influenza A virus (IAV) glycoproteins, was decreased by SERINC5, whether the core was provided by HIV-1, MLV, or M-PMV. In contrast, particles pseudotyped with glycoproteins from M-PMV, parainfluenza virus 5 (PIV5), or rabies virus (RABV) were sensitive to SERINC5, but only with particular retroviral cores. Resistance to SERINC5 did not correlate with reduced SERINC5 incorporation into particles, route of viral entry, or absolute infectivity of the pseudotyped virions. These findings indicate that some non-retroviruses may be sensitive to SERINC5 and that, in addition to the viral glycoprotein, the retroviral core influences sensitivity to SERINC5. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
228. Isolation, Characterization and Targeted Disruption of Mouse Ppia:Cyclophilin A Is Not Essential for Mammalian Cell Viability
- Author
-
Colgan, John, Asmal, Mohammed, and Luban, Jeremy
- Abstract
Cyclophilins (CyPs) are a family of proteins found in organisms ranging from prokaryotes to humans. These molecules exhibit peptidyl-prolyl isomerase activity in vitro,suggesting that they influence the conformation of proteins in cells. CyPs also bind with varying affinities to the immunosuppressive drug cyclosporin A (CsA), a compound used clinically to prevent allograft rejection. The founding member of the family, cyclophilin A (CyPA), is an abundant, ubiquitously expressed protein of unknown function that binds with nanomolar affinity to CsA. Here, we describe the isolation and characterization of mouse Ppia(mPpia), the gene encoding CyPA. Ppiawas isolated using a PCR screen that distinguishes the expressed gene from multiple pseudogenes present in the mouse genome. mPpiaconsists of 5 exons and 4 introns spanning roughly 4.5 kb and maps to chromosome 11 near the centromere. Sequence analysis of a 369-bp fragment from the proximal promoter region of mPpiarevealed the presence of a TATA box and sites recognized by several transcriptional regulators, including Sp1, AP-2, GATA factors, c-Myb, and NF-IL-6. This region is sufficient to drive high-level reporter gene expression in transfected cells. Both copies of Ppiawere disrupted in murine embryonic stem (ES) cells via gene targeting. Ppia−/−ES cells grow normally and differentiate into hematopoeitic precursor cells in vitro,indicating that CyPA is not essential for mammalian cell viability.
- Published
- 2000
- Full Text
- View/download PDF
229. The DHODH inhibitor PTC299 arrests SARS-CoV-2 replication and suppresses induction of inflammatory cytokines.
- Author
-
Luban, Jeremy, Sattler, Rachel A., Mühlberger, Elke, Graci, Jason D., Cao, Liangxian, Weetall, Marla, Trotta, Christopher, Colacino, Joseph M., Bavari, Sina, Strambio-De-Castillia, Caterina, Suder, Ellen L., Wang, Yetao, Soloveva, Veronica, Cintron-Lue, Katherine, Naryshkin, Nikolai A., Pykett, Mark, Welch, Ellen M., O'Keefe, Kylie, Kong, Ronald, and Goodwin, Elizabeth
- Subjects
- *
SARS-CoV-2 , *COVID-19 , *VASCULAR endothelial growth factors , *DIHYDROOROTATE dehydrogenase , *ARREST - Abstract
• COVID-19 is typified by SARS-CoV-2 replication and an excessive inflammatory response. • PTC299 is a small orally bioavailable DHODH inhibitor that blocks pyrimidine synthesis. • PTC299 is a potent inhibitor of the replication of SARS-CoV-2 and other RNA viruses. • PTC299 also reduces the production of cytokines associated with COVID-19 inflammation. • PTC299 may be a promising therapeutic for COVID-19. The coronavirus disease 2019 (COVID-19) pandemic has created an urgent need for therapeutics that inhibit the SARS−COV-2 virus and suppress the fulminant inflammation characteristic of advanced illness. Here, we describe the anti−COVID-19 potential of PTC299, an orally bioavailable compound that is a potent inhibitor of dihydroorotate dehydrogenase (DHODH), the rate-limiting enzyme of the de novo pyrimidine nucleotide biosynthesis pathway. In tissue culture, PTC299 manifests robust, dose-dependent, and DHODH-dependent inhibition of SARS−COV-2 replication (EC 50 range, 2.0–31.6 nM) with a selectivity index >3,800. PTC299 also blocked replication of other RNA viruses, including Ebola virus. Consistent with known DHODH requirements for immunomodulatory cytokine production, PTC299 inhibited the production of interleukin (IL)-6, IL-17A (also called IL-17), IL-17 F, and vascular endothelial growth factor (VEGF) in tissue culture models. The combination of anti-SARS-CoV-2 activity, cytokine inhibitory activity, and previously established favorable pharmacokinetic and human safety profiles render PTC299 a promising therapeutic for COVID-19. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
230. AIM2 regulates anti-tumor immunity and is a viable therapeutic target for melanoma
- Author
-
Fukuda, Keitaro, Okamura, Ken, Riding, Rebecca L., Fan, Xueli, Afshari, Khashayar, Haddadi, Nazgol-Sadat, McCauley, Sean M., Guney, Mehmet H., Luban, Jeremy, Funakoshi, Takeru, Yaguchi, Tomonori, Kawakami, Yutaka, Khvorova, Anastasia, Fitzgerald, Katherine A., and Harris, John E.
- Abstract
The STING and absent in melanoma 2 (AIM2) pathways are activated by the presence of cytosolic DNA, and STING agonists enhance immunotherapeutic responses. Here, we show that dendritic cell (DC) expression of AIM2 within human melanoma correlates with poor prognosis and, in contrast to STING, AIM2 exerts an immunosuppressive effect within the melanoma microenvironment. Vaccination with AIM2-deficient DCs improves the efficacy of both adoptive T cell therapy and anti–PD-1 immunotherapy for “cold tumors,” which exhibit poor therapeutic responses. This effect did not depend on prolonged survival of vaccinated DCs, but on tumor-derived DNA that activates STING-dependent type I IFN secretion and subsequent production of CXCL10 to recruit CD8+ T cells. Additionally, loss of AIM2-dependent IL-1β and IL-18 processing enhanced the treatment response further by limiting the recruitment of regulatory T cells. Finally, AIM2 siRNA-treated mouse DCs in vivo and human DCs in vitro enhanced similar anti-tumor immune responses. Thus, targeting AIM2 in tumor-infiltrating DCs is a promising new treatment strategy for melanoma.
- Published
- 2021
- Full Text
- View/download PDF
231. Single-Cell RNA Profiling Reveals Adipocyte to Macrophage Signaling Sufficient to Enhance Thermogenesis.
- Author
-
Henriques, Felipe, Bedard, Alexander H., Guilherme, Adilson, Kelly, Mark, Chi, Jingyi, Zhang, Peng, Lifshitz, Lawrence M., Bellvé, Karl, Rowland, Leslie A., Yenilmez, Batuhan, Kumar, Shreya, Wang, Yetao, Luban, Jeremy, Weinstein, Lee S., Lin, Jiandie D., Cohen, Paul, and Czech, Michael P.
- Abstract
Adipocytes deficient in fatty acid synthase (iAdFASNKO) emit signals that mimic cold exposure to enhance the appearance of thermogenic beige adipocytes in mouse inguinal white adipose tissues (iWATs). Both cold exposure and iAdFASNKO upregulate the sympathetic nerve fiber (SNF) modulator Neuregulin 4 (Nrg4), activate SNFs, and require adipocyte cyclic AMP/protein kinase A (cAMP/PKA) signaling for beige adipocyte appearance, as it is blocked by adipocyte Gsα deficiency. Surprisingly, however, in contrast to cold-exposed mice, neither iWAT denervation nor Nrg4 loss attenuated adipocyte browning in iAdFASNKO mice. Single-cell transcriptomic analysis of iWAT stromal cells revealed increased macrophages displaying gene expression signatures of the alternately activated type in iAdFASNKO mice, and their depletion abrogated iWAT beiging. Altogether, these findings reveal that divergent cellular pathways are sufficient to cause adipocyte browning. Importantly, adipocyte signaling to enhance alternatively activated macrophages in iAdFASNKO mice is associated with enhanced adipose thermogenesis independent of the sympathetic neuron involvement this process requires in the cold. • Light sheet microscopy reveals nerve fibers abutting beige adipocytes in iWAT • iWAT denervation blocks cold-induced but not adipocyte FASNKO-induced iWAT beiging • cAMP/PKA pathway in adipocytes mediates iWAT beiging in adipocyte FASNKO mice • Macrophages but not innervation are required for beiging in adipocyte FASNKO mice Henriques et al. show an alternative pathway to enhance thermogenesis through an adipocyte cAMP/PKA axis in denervated iWAT. Signals emanating from this pathway generate M2-type macrophages associated with iWAT browning. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
232. Reporter Assays for Ebola Virus Nucleoprotein Oligomerization, Virion-Like Particle Budding, and Minigenome Activity Reveal the Importance of Nucleoprotein Amino Acid Position 111.
- Author
-
Lin, Aaron E., Diehl, William E., Cai, Yingyun, Finch, Courtney L., Akusobi, Chidiebere, Kirchdoerfer, Robert N., Bollinger, Laura, Schaffner, Stephen F., Brown, Elizabeth A., Saphire, Erica Ollmann, Andersen, Kristian G., Kuhn, Jens H., Luban, Jeremy, and Sabeti, Pardis C.
- Subjects
EBOLA virus ,EBOLA virus disease ,OLIGOMERIZATION ,AMINO acids ,PATHOGENIC viruses ,PROTEIN structure - Abstract
For highly pathogenic viruses, reporter assays that can be rapidly performed are critically needed to identify potentially functional mutations for further study under maximal containment (e.g., biosafety level 4 [BSL-4]). The Ebola virus nucleoprotein (NP) plays multiple essential roles during the viral life cycle, yet few tools exist to study the protein under BSL-2 or equivalent containment. Therefore, we adapted reporter assays to measure NP oligomerization and virion-like particle (VLP) production in live cells and further measured transcription and replication using established minigenome assays. As a proof-of-concept, we examined the NP-R111C substitution, which emerged during the 2013–2016 Western African Ebola virus disease epidemic and rose to high frequency. NP-R111C slightly increased NP oligomerization and VLP budding but slightly decreased transcription and replication. By contrast, a synthetic charge-reversal mutant, NP-R111E, greatly increased oligomerization but abrogated transcription and replication. These results are intriguing in light of recent structures of NP oligomers, which reveal that the neighboring residue, K110, forms a salt bridge with E349 on adjacent NP molecules. By developing and utilizing multiple reporter assays, we find that the NP-111 position mediates a complex interplay between NP's roles in protein structure, virion budding, and transcription and replication. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
233. Real-Time Analysis of Individual Ebola Virus Glycoproteins Reveals Pre-Fusion, Entry-Relevant Conformational Dynamics.
- Author
-
Durham, Natasha D., Howard, Angela R., Govindan, Ramesh, Senjobe, Fernando, Fels, J. Maximilian, Diehl, William E., Luban, Jeremy, Chandran, Kartik, and Munro, James B.
- Subjects
EBOLA virus ,FLUORESCENCE resonance energy transfer ,GLYCOPROTEINS ,MEMBRANE proteins ,VIRAL envelope proteins ,STRUCTURAL dynamics ,CONFORMATIONAL analysis - Abstract
The Ebola virus (EBOV) envelope glycoprotein (GP) mediates the fusion of the virion membrane with the membrane of susceptible target cells during infection. While proteolytic cleavage of GP by endosomal cathepsins and binding of the cellular receptor Niemann-Pick C1 protein (NPC1) are essential steps for virus entry, the detailed mechanisms by which these events promote membrane fusion remain unknown. Here, we applied single-molecule Förster resonance energy transfer (smFRET) imaging to investigate the structural dynamics of the EBOV GP trimeric ectodomain, and the functional transmembrane protein on the surface of pseudovirions. We show that in both contexts, pre-fusion GP is dynamic and samples multiple conformations. Removal of the glycan cap and NPC1 binding shift the conformational equilibrium, suggesting stabilization of conformations relevant to viral fusion. Furthermore, several neutralizing antibodies enrich alternative conformational states. This suggests that these antibodies neutralize EBOV by restricting access to GP conformations relevant to fusion. This work demonstrates previously unobserved dynamics of pre-fusion EBOV GP and presents a platform with heightened sensitivity to conformational changes for the study of GP function and antibody-mediated neutralization. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
234. The hidden cost of genetic resistance to HIV-1
- Author
-
Luban, Jeremy
- Abstract
Assessment of more than 400,000 people over the age of 40 demonstrates that homozygosity for a CCR5variant that prevents HIV-1 infection comes at the cost of increased rates of mortality.
- Published
- 2019
- Full Text
- View/download PDF
235. IFIH1 (MDA5) is required for innate immune detection of intron-containing RNA expressed from the HIV-1 provirus.
- Author
-
Guney, Mehmet Hakan, Nagalekshmi, Karthika, McCauley, Sean Matthew, Carbone, Claudia, Aydemir, Ozkan, and Luban, Jeremy
- Subjects
- *
NIPAH virus , *TYPE I interferons , *DENDRITIC cells , *HAIRPIN (Genetics) , *BLOOD cells - Abstract
Intron-containing RNA expressed from the HIV-1 provirus activates type 1 interferon in primary human blood cells, including CD4+ T cells, macrophages, and dendritic cells. To identify the innate immune receptor required for detection of intron-containing RNA expressed from the HIV-1 provirus, a loss-of-function screen was performed with short hairpin RNA-expressing lentivectors targeting twenty-one candidate genes in human monocyte-derived dendritic cells. Among the candidate genes tested, only knockdown of XPO1 (CRM1), IFIH1 (MDA5), or MAVS prevented activation of the interferon-stimulated gene ISG15. The importance of IFIH1 protein was demonstrated by rescue of the knockdown with nontargetable IFIH1 coding sequence. Inhibition of HIV-1-induced ISG15 by the IFIH1-specific Nipah virus V protein, and by IFIH1-transdominant 2-CARD domain-deletion or phosphomimetic point mutations, indicates that IFIH1 (MDA5) filament formation, dephosphorylation, and association with MAVS are all required for innate immune activation in response to HIV-1 transduction. Since both IFIH1 (MDA5) and DDX58 (RIG-I) signal via MAVS, the specificity of HIV-1 RNA detection by IFIH1 was demonstrated by the fact that DDX58 knockdown had no effect on activation. RNA-Seq showed that IFIH1 knockdown in dendritic cells globally disrupted the induction of IFN-stimulated genes by HIV-1. Finally, specific enrichment of unspliced HIV-1 RNA by IFIH1 (MDA5), over two orders of magnitude, was revealed by formaldehyde cross-linking immunoprecipitation (f-CLIP). These results demonstrate that IFIH1 is the innate immune receptor for intron-containing RNA from the HIV-1 provirus and that IFIH1 potentially contributes to chronic inflammation in people living with HIV-1, even in the presence of effective antiretroviral therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
236. MOESM5 of HIV-1 capsid is involved in post-nuclear entry steps
- Author
-
Nan-Yu Chen, Lihong Zhou, Gane, Paul, Opp, Silvana, Ball, Neil, Nicastro, Giuseppe, Zufferey, Madeleine, Buffone, Cindy, Luban, Jeremy, Selwood, David, Diaz-Griffero, Felipe, Taylor, Ian, and Ariberto Fassati
- Subjects
3. Good health - Abstract
Additional file 5: Fig. S5. CsA washout assays in cells depleted of Nup153 or control DsRed cells. T5Cyp cells expressing an shRNA against Nup153 or DsRed (control) were infected at an MOI of 0.01–0.05 with an HIV-1GFP vector (WT) in the presence of CsA (1 μM). The drug was washed out at the indicated time points (time of washout) and cells were analysed by FACS 48 h later to determine the percentage of infected (GFP+) cells. To control for specificity, cells were infected in the same way using the N74D mutant virus. Raw data of three independent experiments are shown. The data have been used to compile fold rescue levels shown in Figure 6.
237. TRIM5α selectively binds a restriction-sensitive retroviral capsid
- Author
-
Sebastian, Sarah and Luban, Jeremy
- Subjects
viruses ,Virology ,FOS: Biological sciences ,Protein binding ,Retroviruses--Enzymes ,Microbiology ,3. Good health - Abstract
TRIM5 is a potent retrovirus inhibitor that targets viruses bearing particular capsid (CA) residues. In most primate species, retroviral restriction requires the C-terminal SPRY domain unique to the α-isoform of TRIM5, but the mechanism by which susceptible viruses are recognized and targeted for restriction is unknown. Here we show that TRIM5α binds retroviral CA from detergent-stripped virions in a SPRY-dependent manner with sufficient discrimination to account for the exquisite specificity of restriction.
238. MOESM4 of HIV-1 capsid is involved in post-nuclear entry steps
- Author
-
Nan-Yu Chen, Lihong Zhou, Gane, Paul, Opp, Silvana, Ball, Neil, Nicastro, Giuseppe, Zufferey, Madeleine, Buffone, Cindy, Luban, Jeremy, Selwood, David, Diaz-Griffero, Felipe, Taylor, Ian, and Ariberto Fassati
- Subjects
3. Good health - Abstract
Additional file 4: Fig. S4. Conditions for the CsA washout assay. (A) H126Q cells were infected with WT HIV-1GFP vector by spinoculation in the presence of CsA (1 μM) or C-A1 (3 μM). The drug was washed out at the indicated time points (time of washout) and cells were analysed by FACS 48 h later to determine the percentage of infected (GFP+) cells. (B) Same as (A) but T5Cyp cells expressing functional human TRIMCyp were used. (C) A prolonged time of uncoating assay, in which CsA was washed out at the indicated time points. Rescue of infection reaches a plateau after 10 h. Average ± SD of three independent experiments are shown in (A-C).
239. Association of TRIM22 with the Type 1 Interferon Response and Viral Control during Primary HIV-1 Infection
- Author
-
Singh, Ravesh, Gaiha, Gaurav, Werner, Lise, McKim, Kevin, Mlisana, Koleka P., Luban, Jeremy, Walker, Bruce D., Abdool Karim, Salim, Brass, Abraham L., and Ndung'u, Thumbi
- Subjects
Epidemiology ,Virology ,3. Good health - Abstract
Type 1 interferons (IFNs) induce the expression of the tripartite interaction motif (TRIM) family of E3 ligases, but the contribution of these antiviral factors to HIV pathogenesis is not completely understood. We hypothesized that the increased expression of select type 1 IFN and TRIM isoforms is associated with a significantly lower likelihood of HIV-1 acquisition and viral control during primary HIV-1 infection. We measured IFN-α, IFN-β, myxovirus resistance protein A (MxA), human TRIM5α (huTRIM5α), and TRIM22 mRNA levels in peripheral blood mononuclear cells (PBMCs) of high-risk, HIV-1-uninfected participants and HIV-1-positive study participants. Samples were available for 32 uninfected subjects and 28 infected persons, all within 1 year of infection. HIV-1-positive participants had higher levels of IFN-β (P = 0.0005), MxA (P = 0.007), and TRIM22 (P = 0.01) and lower levels of huTRIM5α (P < 0.001) than did HIV-1-negative participants. TRIM22 but not huTRIM5α correlated positively with type 1 IFN (IFN-α, IFN-β, and MxA) (all P < 0.0001). In a multivariate model, increased MxA expression showed a significant positive association with viral load (P = 0.0418). Furthermore, TRIM22 but not huTRIM5α, IFN-α, IFN-β, or MxA showed a negative correlation with plasma viral load (P = 0.0307) and a positive correlation with CD4+ T-cell counts (P = 0.0281). In vitro studies revealed that HIV infection induced TRIM22 expression in PBMCs obtained from HIV-negative donors. Stable TRIM22 knockdown resulted in increased HIV-1 particle release and replication in Jurkat reporter cells. Collectively, these data suggest concordance between type 1 IFN and TRIM22 but not huTRIM5α expression in PBMCs and that TRIM22 likely acts as an antiviral effector in vivo.
240. MOESM5 of HIV-1 capsid is involved in post-nuclear entry steps
- Author
-
Nan-Yu Chen, Lihong Zhou, Gane, Paul, Opp, Silvana, Ball, Neil, Nicastro, Giuseppe, Zufferey, Madeleine, Buffone, Cindy, Luban, Jeremy, Selwood, David, Diaz-Griffero, Felipe, Taylor, Ian, and Ariberto Fassati
- Subjects
3. Good health - Abstract
Additional file 5: Fig. S5. CsA washout assays in cells depleted of Nup153 or control DsRed cells. T5Cyp cells expressing an shRNA against Nup153 or DsRed (control) were infected at an MOI of 0.01–0.05 with an HIV-1GFP vector (WT) in the presence of CsA (1 μM). The drug was washed out at the indicated time points (time of washout) and cells were analysed by FACS 48 h later to determine the percentage of infected (GFP+) cells. To control for specificity, cells were infected in the same way using the N74D mutant virus. Raw data of three independent experiments are shown. The data have been used to compile fold rescue levels shown in Figure 6.
241. MOESM4 of HIV-1 capsid is involved in post-nuclear entry steps
- Author
-
Nan-Yu Chen, Lihong Zhou, Gane, Paul, Opp, Silvana, Ball, Neil, Nicastro, Giuseppe, Zufferey, Madeleine, Buffone, Cindy, Luban, Jeremy, Selwood, David, Diaz-Griffero, Felipe, Taylor, Ian, and Ariberto Fassati
- Subjects
3. Good health - Abstract
Additional file 4: Fig. S4. Conditions for the CsA washout assay. (A) H126Q cells were infected with WT HIV-1GFP vector by spinoculation in the presence of CsA (1 μM) or C-A1 (3 μM). The drug was washed out at the indicated time points (time of washout) and cells were analysed by FACS 48 h later to determine the percentage of infected (GFP+) cells. (B) Same as (A) but T5Cyp cells expressing functional human TRIMCyp were used. (C) A prolonged time of uncoating assay, in which CsA was washed out at the indicated time points. Rescue of infection reaches a plateau after 10 h. Average ± SD of three independent experiments are shown in (A-C).
242. The retroviral restriction factor TRIM5α
- Author
-
Sebastian, Sarah, Luban, Jeremy, Sebastian, Sarah, and Luban, Jeremy
- Abstract
Retroviruses are obligate intracellular parasites that have coevolved with their hosts for millions of years. It is therefore not surprising that retroviruses take advantage of numerous host factors during their life cycle. In addition to positive cellular factors that are of use to the virus, host cells have also evolved intracellular proteins to antagonize the retroviral replication cycle. Such inhibitory cellular factors have been called retroviral restriction factors. Recently, several such restriction factors have been cloned, including Friend virus susceptibility factor 1, apolipoprotein B mRNA-editing enzyme catalytic proteins 3F and 3G, and ZAP. Here, we review the explosion of publications from the past 2 years concerning TRIM5, a host factor that potently inhibits HIV-1 and other retroviruses
243. Cyclosporine A-sensitive, cyclophilin B-dependent endoplasmic reticulum-associated degradation
- Author
-
Bernasconi, Riccardo, Soldà, Tatiana, Galli, Carmela, Pertel, Thomas, Luban, Jeremy, Molinari, Maurizio, Bernasconi, Riccardo, Soldà, Tatiana, Galli, Carmela, Pertel, Thomas, Luban, Jeremy, and Molinari, Maurizio
- Abstract
Peptidyl-prolyl cis/trans isomerases (PPIs) catalyze cis/trans isomerization of peptide bonds preceding proline residues. The involvement of PPI family members in protein refolding has been established in test tube experiments. Surprisingly, however, no data is available on the involvement of endoplasmic reticulum (ER)-resident members of the PPI family in protein folding, quality control or disposal in the living cell. Here we report that the immunosuppressive drug cyclosporine A (CsA) selectively inhibits the degradation of a subset of misfolded proteins generated in the ER. We identify cyclophilin B (CyPB) as the ER-resident target of CsA that catalytically enhances disposal from the ER of ERAD-LS substrates containing cis proline residues. Our manuscript presents the first evidence for enzymatic involvement of a PPI in protein quality control in the ER of living cells.
244. Intron-containing RNA from the HIV-1 provirus activates type I interferon and inflammatory cytokines.
- Author
-
McCauley, Sean Matthew, Kim, Kyusik, Nowosielska, Anetta, Dauphin, Ann, Yurkovetskiy, Leonid, Diehl, William Edward, and Luban, Jeremy
- Abstract
HIV-1-infected people who take drugs that suppress viremia to undetectable levels are protected from developing AIDS. Nonetheless, HIV-1 establishes proviruses in long-lived CD4
+ memory T cells, and perhaps other cell types, that preclude elimination of the virus even after years of continuous antiviral therapy. Here we show that the HIV-1 provirus activates innate immune signaling in isolated dendritic cells, macrophages, and CD4+ T cells. Immune activation requires transcription from the HIV-1 provirus and expression of CRM1-dependent, Rev-dependent, RRE-containing, unspliced HIV-1 RNA. If rev is provided in trans, all HIV-1 coding sequences are dispensable for activation except those cis-acting sequences required for replication or splicing. Our results indicate that the complex, post-transcriptional regulation intrinsic to HIV-1 RNA is detected by the innate immune system as a danger signal, and that drugs which disrupt HIV-1 transcription or HIV-1 RNA metabolism would add qualitative benefit to current antiviral drug regimens. During HIV infection, antiviral therapy can suppress viraemia to undetectable levels and hinder the progression towards AIDS; however the HIV-1 provirus can remain in long-lived CD4+ memory T cells. Here the authors show that intronic RNA from the HIV-1 provirus can induce type I interferon and inflammatory cytokine production. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
245. Retraction Note: The hidden cost of genetic resistance to HIV-1
- Author
-
Luban, Jeremy
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2019
- Full Text
- View/download PDF
246. TRIM5α Restricts Flavivirus Replication by Targeting the Viral Protease for Proteasomal Degradation
- Author
-
Chiramel, Abhilash I., Meyerson, Nicholas R., McNally, Kristin L., Broeckel, Rebecca M., Montoya, Vanessa R., Méndez-Solís, Omayra, Robertson, Shelly J., Sturdevant, Gail L., Lubick, Kirk J., Nair, Vinod, Youseff, Brian H., Ireland, Robin M., Bosio, Catharine M., Kim, Kyusik, Luban, Jeremy, Hirsch, Vanessa M., Taylor, R. Travis, Bouamr, Fadila, Sawyer, Sara L., and Best, Sonja M.
- Abstract
Tripartite motif-containing protein 5α (TRIM5α) is a cellular antiviral restriction factor that prevents early events in retrovirus replication. The activity of TRIM5α is thought to be limited to retroviruses as a result of highly specific interactions with capsid lattices. In contrast to this current understanding, we show that both human and rhesus macaque TRIM5α suppress replication of specific flaviviruses. Multiple viruses in the tick-borne encephalitis complex are sensitive to TRIM5α-dependent restriction, but mosquito-borne flaviviruses, including yellow fever, dengue, and Zika viruses, are resistant. TRIM5α suppresses replication by binding to the viral protease NS2B/3 to promote its K48-linked ubiquitination and proteasomal degradation. Importantly, TRIM5α contributes to the antiviral function of IFN-I against sensitive flaviviruses in human cells. Thus, TRIM5α possesses remarkable plasticity in the recognition of diverse virus families, with the potential to influence human susceptibility to emerging flaviviruses of global concern.
- Published
- 2019
- Full Text
- View/download PDF
247. Comparative Analysis of Immune Cells Reveals a Conserved Regulatory Lexicon
- Author
-
Donnard, Elisa, Vangala, Pranitha, Afik, Shaked, McCauley, Sean, Nowosielska, Anetta, Kucukural, Alper, Tabak, Barbara, Zhu, Xiaopeng, Diehl, William, McDonel, Patrick, Yosef, Nir, Luban, Jeremy, and Garber, Manuel
- Abstract
Most well-characterized enhancers are deeply conserved. In contrast, genome-wide comparative studies of steady-state systems showed that only a small fraction of active enhancers are conserved. To better understand conservation of enhancer activity, we used a comparative genomics approach that integrates temporal expression and epigenetic profiles in an innate immune system. We found that gene expression programs diverge among mildly induced genes, while being highly conserved for strongly induced genes. The fraction of conserved enhancers varies greatly across gene expression programs, with induced genes and early-response genes, in particular, being regulated by a higher fraction of conserved enhancers. Clustering of conserved accessible DNA sequences within enhancers resulted in over 60 sequence motifs including motifs for known factors, as well as many with unknown function. We further show that the number of instances of these motifs is a strong predictor of the responsiveness of a gene to pathogen detection.
- Published
- 2018
- Full Text
- View/download PDF
248. More than one way to TRIM a capsid.
- Author
-
Luban, Jeremy
- Subjects
- *
CAPSIDS , *ADENOVIRUSES , *BIODEGRADATION , *PROTEASOMES , *VIRAL antibodies , *CYTOPLASM - Abstract
The article focuses on tripartite motif-containing protein 21 (TRIM21) which is found to degrade virion capsid. The author states that the TRIM21 binds the adenovirus capsid in the cytoplasm and triggers capsid degradation by the proteasome. He note that neutralizing antibodies are believed to block interaction of the virion with cognate cell surface receptors on susceptible target cells.
- Published
- 2012
- Full Text
- View/download PDF
249. Regulation of Ebola GP conformation and membrane binding by the chemical environment of the late endosome.
- Author
-
Jain, Aastha, Govindan, Ramesh, Berkman, Alex R., Luban, Jeremy, Díaz-Salinas, Marco A., Durham, Natasha D., and Munro, James B.
- Subjects
- *
FLUORESCENCE resonance energy transfer , *EBOLA virus disease , *EBOLA virus , *VIRAL envelope proteins , *CALCIUM ions , *CELL membranes - Abstract
Interaction between the Ebola virus envelope glycoprotein (GP) and the endosomal membrane is an essential step during virus entry into the cell. Acidic pH and Ca2+ have been implicated in mediating the GP-membrane interaction. However, the molecular mechanism by which these environmental factors regulate the conformational changes that enable engagement of GP with the target membrane is unknown. Here, we apply fluorescence correlation spectroscopy (FCS) and single-molecule Förster resonance energy transfer (smFRET) imaging to elucidate how the acidic pH, Ca2+ and anionic phospholipids in the late endosome promote GP-membrane interaction, thereby facilitating virus entry. We find that bis(monoacylglycero)phosphate (BMP), which is specific to the late endosome, is especially critical in determining the Ca2+-dependence of the GP-membrane interaction. Molecular dynamics (MD) simulations suggested residues in GP that sense pH and induce conformational changes that make the fusion loop available for insertion into the membrane. We similarly confirm residues in the fusion loop that mediate GP's interaction with Ca2+, which likely promotes local conformational changes in the fusion loop and mediates electrostatic interactions with the anionic phospholipids. Collectively, our results provide a mechanistic understanding of how the environment of the late endosome regulates the timing and efficiency of virus entry. Author summary: Ebola virus causes disease in humans with high fatality. A better understanding of how Ebola virus enters cells is critical to inform the development of novel therapeutic and preventative measures. The viral glycoprotein present on the surface of the virus mediates attachment to cells and subsequent entry through a poorly understood mechanism involving fusion of viral and cellular membranes. Here, we employ computational and experimental biophysical techniques to understand how the Ebola glycoprotein senses chemical cues in its environment, such as pH, calcium ions, and specific lipid species to ensure that entry occurs at the right time and place. Our results specify elements of the glycoprotein that control its structure under changing physiological environments. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
250. Emerging role of cyclophilin A in HIV-1 infection: from producer cell to the target cell nucleus.
- Author
-
Padron, Adrian, Prakash, Prem, Pandhare, Jui, Luban, Jeremy, Aiken, Chris, Balasubramaniam, Muthukumar, and Dash, Chandravanu
- Subjects
- *
CELL nuclei , *HIV , *CYCLOPHILINS , *CYTOSKELETAL proteins , *KNOWLEDGE gap theory - Abstract
The HIV-1 genome encodes a small number of proteins with structural, enzymatic, regulatory, and accessory functions. These viral proteins interact with a number of host factors to promote the early and late stages of HIV-1 infection. During the early stages of infection, interactions between the viral proteins and host factors enable HIV-1 to enter the target cell, traverse the cytosol, dock at the nuclear pore, gain access to the nucleus, and integrate into the host genome. Similarly, the viral proteins recruit another set of host factors during the late stages of infection to orchestrate HIV-1 transcription, translation, assembly, and release of progeny virions. Among the host factors implicated in HIV-1 infection, Cyclophilin A (CypA) was identified as the first host factor to be packaged within HIV-1 particles. It is now well established that CypA promotes HIV-1 infection by directly binding to the viral capsid. Mechanistic models to pinpoint CypA's role have spanned from an effect in the producer cell to the early steps of infection in the target cell. In this review, we will describe our understanding of the role(s) of CypA in HIV-1 infection, highlight the current knowledge gaps, and discuss the potential role of this host factor in the post-nuclear entry steps of HIV-1 infection. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.