Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Infect Dis ; 2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38537250

RESUMEN

Concerns regarding toxicity and resistance of current drugs have been reported in visceral leishmaniasis. Anti-microbial peptides are considered as new promising candidates and amongst them, human cathelicidin hCAP18/LL-37 showed significant parasite killing on drug-sensitive and resistant Leishmania promastigotes, coupled with its apoptosis-inducing role. Administration of hCAP18/LL-37 in infected macrophages also decreased parasite survival and increased the host favorable cytokine IL-12. However, 1,25-dihydroxyvitamin D3 (VitD3)-induced endogenous hCAP18/LL-37 production was hampered in infected THP-1 cells. Infection also suppressed the VitD3-receptor (VDR), transcription factor of hCAP18/LL-37. cAMP response element modulator (CREM), the repressor of VDR, was induced in infection resulting in suppression of both VDR and cathelicidin expression. PGE2/cAMP/PKA axis was found to regulate CREM induction during infection and silencing CREM in infected cells and BALB/c mice led to decreased parasite survival. Present study thus documents the anti-leishmanial potential of cathelicidin and further identifies CREM as a repressor of cathelicidin in Leishmania infection.

2.
J Immunol ; 208(11): 2540-2548, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35562118

RESUMEN

In the early phase of infection, the intramacrophage pathogen Leishmania donovani protects its niche with the help of the antiapoptotic protein myeloid cell leukemia-1 (MCL-1). Whether Leishmania could exploit MCL-1, an extremely labile protein, at the late phase is still unclear. A steady translational level of MCL-1 observed up to 48 h postinfection and increased caspase-3 activity in MCL-1-silenced infected macrophages documented its importance in the late hours of infection. The transcript level of MCL-1 showed a sharp decline at 6 h postinfection, and persistent MCL-1 expression in cyclohexamide-treated cells negates the possibility of de novo protein synthesis, thereby suggesting infection-induced stability. Increased ubiquitination, a prerequisite for proteasomal degradation of MCL-1, was also found to be absent in the late hours of infection. Lack of interaction with its specific E3 ubiquitin ligase MULE (MCL-1 ubiquitin ligase E3) and specific deubiquitinase USP9X prompted us to search for blockade of the ubiquitin-binding site in MCL-1. To this end, TCTP (translationally controlled tumor protein), a well-known binding partner of MCL-1 and antiapoptotic regulator, was found to be strongly associated with MCL-1 during infection. Phosphorylation of TCTP, a requirement for MCL-1 binding, was also increased in infected macrophages. Knockdown of TCTP decreased MCL-1 expression and short hairpin RNA-mediated silencing of TCTP in an infected mouse model of visceral leishmaniasis showed decreased parasite burden and induction of liver cell apoptosis. Collectively, our investigation revealed a key mechanism of how L. donovani exploits TCTP to establish infection within the host.


Asunto(s)
Leishmania donovani , Leishmaniasis Visceral , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteína Tumoral Controlada Traslacionalmente 1 , Animales , Proteínas Reguladoras de la Apoptosis , Macrófagos/parasitología , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Proteína Tumoral Controlada Traslacionalmente 1/metabolismo , Ubiquitina-Proteína Ligasas
3.
J Virol ; 95(13): e0022921, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-33883219

RESUMEN

SERINC5 restricts nef-defective HIV-1 by affecting early steps of the virus life cycle. Distantly related retroviruses with a wide host range encode virulent factors in response to challenge by SERINC5. However, the evolutionary origins of this antiretroviral activity, its prevalence among the paralogs, and its ability to target retroviruses remain understudied. In agreement with previous studies, we found that four human SERINC paralogs inhibit nef-defective HIV-1, with SERINC2 being an exception. Here, we demonstrate that this lack of activity in human SERINC2 is associated with its post-whole-genome duplication (post-WGD) divergence, as evidenced by the ability of pre-WGD orthologs from Saccharomyces cerevisiae and flies and a post-WGD-proximate SERINC2 from coelacanths to inhibit the virus. Intriguingly, Nef is unable to counter coelacanth SERINC2, indicating that such activity was directed toward other retroviruses found in coelacanths (like foamy viruses). However, foamy virus-derived vectors are intrinsically resistant to the action of SERINC2, and we show that the foamy virus envelope confers this resistance by affecting its steady-state levels. Our study highlights an ancient origin of antiretroviral activity in SERINCs and a hitherto-unknown interaction with a foamy virus. IMPORTANCESERINC5 constitutes a critical barrier to the propagation of retroviruses, as highlighted by parallel emergence of anti-SERINC5 activities among distant retroviral lineages. Therefore, understanding the origin and evolution of these host factors will provide key information about virus-host relationships that can be exploited for future drug development. Here, we show that SERINC5-mediated nef-defective HIV-1 infection inhibition is evolutionarily conserved. SERINC2 from coelacanth restricts HIV-1, and it was functionally adapted to target foamy viruses. Our findings provide insights into the evolutionary origin of antiretroviral activity in the SERINC gene family and uncover the role of SERINCs in shaping the long-term conflicts between retroviruses and their hosts.


Asunto(s)
VIH-1/crecimiento & desarrollo , Proteínas de la Membrana/metabolismo , Spumavirus/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Replicación Viral/fisiología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Animales , Antirretrovirales/metabolismo , Línea Celular , Peces/genética , Células HEK293 , Haplorrinos/genética , Humanos , Proteínas de la Membrana/genética , Saccharomyces cerevisiae/metabolismo
4.
Nature ; 526(7572): 212-7, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26416734

RESUMEN

HIV-1 Nef, a protein important for the development of AIDS, has well-characterized effects on host membrane trafficking and receptor downregulation. By an unidentified mechanism, Nef increases the intrinsic infectivity of HIV-1 virions in a host-cell-dependent manner. Here we identify the host transmembrane protein SERINC5, and to a lesser extent SERINC3, as a potent inhibitor of HIV-1 particle infectivity that is counteracted by Nef. SERINC5 localizes to the plasma membrane, where it is efficiently incorporated into budding HIV-1 virions and impairs subsequent virion penetration of susceptible target cells. Nef redirects SERINC5 to a Rab7-positive endosomal compartment and thereby excludes it from HIV-1 particles. The ability to counteract SERINC5 was conserved in Nef encoded by diverse primate immunodeficiency viruses, as well as in the structurally unrelated glycosylated Gag from murine leukaemia virus. These examples of functional conservation and convergent evolution emphasize the fundamental importance of SERINC5 as a potent anti-retroviral factor.


Asunto(s)
VIH-1/fisiología , Interacciones Huésped-Patógeno , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Virión/química , Virión/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Línea Celular , Membrana Celular/metabolismo , Membrana Celular/virología , Endosomas/química , Endosomas/metabolismo , Evolución Molecular , Productos del Gen gag/metabolismo , Productos del Gen nef/química , Productos del Gen nef/metabolismo , VIH-1/química , Especificidad del Huésped , Humanos , Virus de la Leucemia Murina/química , Virus de la Leucemia Murina/fisiología , Glicoproteínas de Membrana , Proteínas de la Membrana/análisis , Proteínas de Neoplasias/metabolismo , Primates/virología , Receptores de Superficie Celular/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
5.
Proc Natl Acad Sci U S A ; 113(46): 13197-13202, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27803322

RESUMEN

The lentivirus equine infectious anemia virus (EIAV) encodes the small protein S2, a pathogenic determinant that is important for virus replication and disease progression in horses. No molecular function had been linked to this accessory protein. We report that S2 can replace the activity of Negative factor (Nef) in HIV-1 infectivity, being required to antagonize the inhibitory activity of Serine incorporator (SERINC) proteins on Nef-defective HIV-1. Like Nef, S2 excludes SERINC5 from virus particles and requires an ExxxLL motif predicted to recruit the clathrin adaptor, Adaptor protein 2 (AP2). Accordingly, functional endocytic machinery is essential for S2-mediated infectivity enhancement, and S2-mediated enhancement is impaired by inhibitors of clathrin-mediated endocytosis. In addition to retargeting SERINC5 to a late endosomal compartment, S2 promotes host factor degradation. Emphasizing the similarity with Nef, we show that S2 is myristoylated, and, as is compatible with a crucial role in posttranslational modification, its N-terminal glycine is required for anti-SERINC5 activity. EIAV-derived vectors devoid of S2 are less susceptible than HIV-1 to the inhibitory effect of both human and equine SERINC5. We then identified the envelope glycoprotein of EIAV as a determinant that also modulates retroviral susceptibility to SERINC5, indicating that EIAV has a bimodal ability to counteract the host factor. S2 shares no sequence homology with other retroviral factors known to counteract SERINC5. Like the primate lentivirus Nef and the gammaretrovirus glycoGag, the accessory protein from EIAV is an example of a retroviral virulence determinant that independently evolved SERINC5-antagonizing activity. SERINC5 therefore plays a critical role in the interaction of the host with diverse retrovirus pathogens.


Asunto(s)
Proteínas de la Membrana , Proteínas de Neoplasias , Receptores de Superficie Celular , Proteínas Virales , Factores de Virulencia , Línea Celular , VIH-1/fisiología , Humanos , Virus de la Anemia Infecciosa Equina , Glicoproteínas de Membrana , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Replicación Viral
6.
J Biol Chem ; 292(14): 6014-6026, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28179429

RESUMEN

The host proteins, SERINC3 and SERINC5, have been recently shown to incorporate into HIV-1 particles and compromise their ability to fuse with target cells, an effect that is antagonized by the viral Nef protein. Envelope (Env) glycoproteins from different HIV-1 isolates exhibit a broad range of sensitivity to SERINC-mediated restriction, and the mechanism by which SERINCs interfere with HIV-1 fusion remains unclear. Here, we show that incorporation of SERINC5 into virions in the absence of Nef inhibits the formation of small fusion pores between viruses and cells. Strikingly, we found that SERINC5 promotes spontaneous functional inactivation of sensitive but not resistant Env glycoproteins. Although SERINC5-Env interaction was not detected by co-immunoprecipitation, incorporation of this protein enhanced the exposure of the conserved gp41 domains and sensitized the virus to neutralizing antibodies and gp41-derived inhibitory peptides. These results imply that SERINC5 restricts HIV-1 fusion at a step prior to small pore formation by selectively inactivating sensitive Env glycoproteins, likely through altering their conformation. The increased HIV-1 sensitivity to anti-gp41 antibodies and peptides suggests that SER5 also delays refolding of the remaining fusion-competent Env trimers.


Asunto(s)
Proteína gp41 de Envoltorio del VIH/metabolismo , VIH-1/metabolismo , Proteínas de la Membrana/metabolismo , Replegamiento Proteico , Células HEK293 , Anticuerpos Anti-VIH/farmacología , Proteína gp41 de Envoltorio del VIH/genética , VIH-1/genética , Humanos , Proteínas de la Membrana/genética , Dominios Proteicos , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo
7.
Angew Chem Int Ed Engl ; 57(20): 5735-5739, 2018 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-29573526

RESUMEN

Reported is the first organocatalytic asymmetric 1,3-alkyl shift in alkyl aryl ethers for the synthesis of chiral 3,3'-diaryloxindoles using a chiral Brønsted acid catalyst. Preliminary results showed that each enantiomer of the 3,3'-diaryloxindole, and a racemic mixture, showed different antiproliferative activities against HeLa cell lines by using an MTT assay.


Asunto(s)
Éteres/química , Oxindoles/síntesis química , Ácidos Fosfóricos/química , Catálisis , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Células HeLa , Humanos , Modelos Moleculares , Estructura Molecular , Oxindoles/química , Oxindoles/farmacología , Estereoisomerismo
8.
J Immunol ; 193(7): 3644-53, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25187664

RESUMEN

CD40 plays dual immunoregulatory roles in Leishmania major infection and tumor regression. The functional duality emerges from CD40-induced reciprocal p38MAPK and ERK-1/2 phosphorylations. Because phosphotyrosine-based signaling in hematopoietic cells is regulated by the phosphotyrosine phosphatase SHP-1, which is not implied in CD40 signaling, we examined whether SHP-1 played any roles in CD40-induced reciprocal signaling and anti-leishmanial function. We observed that a weaker CD40 stimulation increased SHP-1 activation. ERK-1/2 inhibition or p38MAPK overexpression inhibited CD40-induced SHP-1 activation. An ultra-low-dose, CD40-induced p38MAPK phosphorylation was enhanced by SHP-1 inhibition but reduced by SHP-1 overexpression. A reverse profile was observed with ERK-1/2 phosphorylation. SHP-1 inhibition reduced syk phosphorylation but increased lyn phosphorylation; syk inhibition reduced but lyn inhibition enhanced CD40-induced SHP-1 phosphorylation. Corroborating these findings, in L. major-infected macrophages, CD40-induced SHP-1 phosphorylation increased and SHP-1 inhibition enhanced CD40-induced p38MAPK activation and inducible NO synthase expression. IL-10 enhanced SHP-1 phosphorylation and CD40-induced ERK-1/2 phosphorylation but reduced the CD40-induced p38MAPK phosphorylation, whereas anti-IL-10 Ab exhibited reverse effects on these CD40-induced functions, identifying IL-10 as a crucial element in the SHP-1-MAPK feedback system. Lentivirally overexpressed SHP-1 rendered resistant C57BL/6 mice susceptible to the infection. Lentivirally expressed SHP-1 short hairpin RNA enhanced the CD40-induced L. major parasite killing in susceptible BALB/c mice. Thus, we establish an SHP-1-centered feedback system wherein SHP-1 modulates CD40-induced p38MAPK activation threshold and reciprocal ERK-1/2 activation, establishing itself as a critical regulator of CD40 signaling reciprocity and mechanistically re-emphasizing its role as a potential target against the diseases where CD40 is involved.


Asunto(s)
Antígenos CD40/inmunología , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Macrófagos/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Animales , Activación Enzimática/inmunología , Interleucina-10/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Leishmaniasis Cutánea/patología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Proteína Quinasa 3 Activada por Mitógenos/inmunología , Óxido Nítrico Sintasa de Tipo II , Fosforilación/inmunología , Proteínas Tirosina Quinasas/inmunología , Quinasa Syk , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
9.
FASEB J ; 28(4): 1756-68, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24391131

RESUMEN

Intramacrophage pathogen Leishmania donovani escapes host immune response by subverting Toll-like receptor (TLR) signaling, which is critically regulated by protein ubiquitination. In the present study, we identified tumor necrosis factor receptor-associated factor (TRAF) 3, degradative ubiquitination of which is essential for TLR4 activation, as a target for Leishmania to deactivate LPS-mediated TLR4 signaling. We used LPS-treated RAW 264.7 cells and compared the TLR4-mediated immune response in these cells with L. donovani and L. donovani + LPS costimulated macrophages. TRAF3, which was ubiquitinated (2.1-fold over control) at lys 48 position and subsequently degraded following LPS treatment, persisted in L. donovani and L. donovani + LPS costimulated cells due to defective lys 48 ubiquitination. Lys 63-linked ubiquitination of upstream proteins in the cascade (cIAP1/2 and TRAF6), mandatory for TRAF3 degradation, was also reduced postinfection. This may be attributed to reduced association between ubiquitin-conjugating enzyme Ubc13 and TRAF6 during infection. Inhibition of TRAF3 before infection by shRNA in Balb/c mice showed enhanced IL-12 and TNF-α (10.8- and 8.1-fold over infected control) and decreased spleen parasite burden (61.3% suppression, P<0.001), thereby marking reduction in disease progression. Our findings identified TRAF3 as a novel molecular regulator exploited by Leishmania for successful infection.


Asunto(s)
Leishmania donovani/inmunología , Macrófagos/inmunología , Transducción de Señal/inmunología , Factor 3 Asociado a Receptor de TNF/inmunología , Receptor Toll-Like 4/inmunología , Animales , Línea Celular , Células Cultivadas , Expresión Génica/inmunología , Interacciones Huésped-Parásitos/inmunología , Immunoblotting , Interleucina-12/genética , Interleucina-12/inmunología , Interleucina-12/metabolismo , Leishmania donovani/fisiología , Leishmaniasis Visceral/genética , Leishmaniasis Visceral/inmunología , Leishmaniasis Visceral/parasitología , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Lisina/inmunología , Lisina/metabolismo , Macrófagos/metabolismo , Macrófagos/parasitología , Ratones , Ratones Endogámicos BALB C , Modelos Inmunológicos , FN-kappa B/genética , FN-kappa B/inmunología , FN-kappa B/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitinación/efectos de los fármacos , Ubiquitinación/inmunología
10.
Plasmid ; 71: 1-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24325878

RESUMEN

Lentiviral vector (LV) mediated gene transfer holds great promise to develop stable cell lines for sustained transgene expression providing a valuable alternative to the conventional plasmid transfection based recombinant protein production methods. We report here making a third generation HIV-2 derived LV containing erythropoietin (EPO) gene expression cassette to generate a stable HEK293 cell line secreting EPO constitutively. A high producer cell clone was obtained by limiting dilution and was adapted to serum free medium. The suspension adapted cell clone stably produced milligram per liter quantities of EPO. Subsequent host metabolic engineering using lentiviral RNAi targeted to block an endogenous candidate protease elastase, identified through an in silico approach, resulted in appreciable augmentation of EPO expression above the original level. This study of LV based improved glycoprotein expression with host cell metabolic engineering for stable production of protein therapeutics thus exemplifies the versatility of LV and is of significant future biopharmaceutical importance.


Asunto(s)
Eritropoyetina/biosíntesis , Vectores Genéticos , Lentivirus/genética , Elastasa Pancreática/metabolismo , ARN Interferente Pequeño/metabolismo , Clonación Molecular , Regulación hacia Abajo , Epoetina alfa , Eritropoyetina/genética , Células HEK293 , Humanos , Lentivirus/aislamiento & purificación , Elastasa Pancreática/genética , Plásmidos/genética , Plásmidos/metabolismo , Interferencia de ARN/fisiología , ARN Interferente Pequeño/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Transfección , Transgenes
11.
J Immunol ; 189(2): 924-34, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22685311

RESUMEN

TLRs, which form an interface between mammalian host and microbe, play a key role in pathogen recognition and initiation of proinflammatory response thus stimulating antimicrobial activity and host survival. However, certain intracellular pathogens such as Leishmania can successfully manipulate the TLR signaling, thus hijacking the defensive strategies of the host. Despite the presence of lipophosphoglycan, a TLR2 ligand capable of eliciting host-defensive cytokine response, on the surface of Leishmania, the strategies adopted by the parasite to silence the TLR2-mediated proinflammatory response is not understood. In this study, we showed that Leishmania donovani modulates the TLR2-mediated pathway in macrophages through inhibition of the IKK-NF-κB cascade and suppression of IL-12 and TNF-α production. This may be due to impairment of the association of TRAF6 with the TAK-TAB complex, thus inhibiting the recruitment of TRAF6 in TLR2 signaling. L. donovani infection drastically reduced Lys 63-linked ubiquitination of TRAF6, and the deubiquitinating enzyme A20 was found to be significantly upregulated in infected macrophages. Small interfering RNA-mediated silencing of A20 restored the Lys 63-linked ubiquitination of TRAF6 as well as IL-12 and TNF-α levels with a concomitant decrease in IL-10 and TGF-ß synthesis in infected macrophages. Knockdown of A20 led to lower parasite survival within macrophages. Moreover, in vivo silencing of A20 by short hairpin RNA in BALB/c mice led to increased NF-κB DNA binding and host-protective proinflammatory cytokine response resulting in effective parasite clearance. These results suggest that L. donovani might exploit host A20 to inhibit the TLR2-mediated proinflammatory gene expression, thus escaping the immune responses of the host.


Asunto(s)
Cisteína Endopeptidasas/fisiología , Regulación hacia Abajo/inmunología , Glicoesfingolípidos/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Leishmania donovani/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 2/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Animales , Línea Celular , Femenino , Glicoesfingolípidos/antagonistas & inhibidores , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Leishmania donovani/patogenicidad , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Macrófagos/enzimología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Factor 6 Asociado a Receptor de TNF/antagonistas & inhibidores , Factor 6 Asociado a Receptor de TNF/metabolismo , Receptor Toll-Like 2/fisiología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa
12.
Methods Mol Biol ; 2807: 31-43, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743219

RESUMEN

RNA fluorescence in situ hybridization (FISH) serves as a method for visualizing specific RNA molecules within cells. Its primary utility lies in the observation of messenger RNA (mRNA) molecules associated with particular genes of significance. This technique can also be applied to examine viral transcription and the localization of said transcripts within infected cells. In this context, we provide a comprehensive protocol for the detection, localization, and quantification of HIV-1 transcripts in mammalian cell lines. This encompasses the preparation of required reagents, cellular treatments, visualization, and the subsequent analysis of the data acquired. These parameters play a pivotal role in enhancing our comprehension of the molecular processes during infection, particularly at the crucial transcription phase of the viral life cycle.


Asunto(s)
VIH-1 , Hibridación Fluorescente in Situ , ARN Viral , Transcripción Genética , Hibridación Fluorescente in Situ/métodos , Humanos , ARN Viral/genética , VIH-1/genética , ARN Mensajero/genética , Infecciones por VIH/virología , Línea Celular
13.
Int Immunopharmacol ; 134: 112100, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38728877

RESUMEN

The parasite Leishmania resides as amastigotes within the macrophage parasitophorous vacuoles inflicting the disease Leishmaniasis. Leishmania selectively modulates mitogen-activated protein kinase (MAPK) phosphorylation subverting CD40-triggered anti-leishmanial functions of macrophages. The mechanism of any pathogen-derived molecule induced host MAPK modulation remains poorly understood. Herein, we show that of the fifteen MAPKs, LmjMAPK4 expression is higher in virulent L. major. LmjMAPK4- detected in parasitophorous vacuoles and cytoplasm- binds MEK-1/2, but not MKK-3/6. Lentivirally-overexpressed LmjMAPK4 augments CD40-activated MEK-1/2-ERK-1/2-MKP-1, but inhibits MKK3/6-p38MAPK-MKP-3, phosphorylation. A rationally-identified LmjMAPK4 inhibitor reinstates CD40-activated host-protective anti-leishmanial functions in L. major-infected susceptible BALB/c mice. These results identify LmjMAPK4 as a MAPK modulator at the host-pathogen interface and establish a pathogen-intercepted host receptor signaling as a scientific rationale for identifying drug targets.


Asunto(s)
Antígenos CD40 , Leishmania major , Leishmaniasis Cutánea , Macrófagos , Ratones Endogámicos BALB C , Transducción de Señal , Animales , Leishmania major/inmunología , Leishmania major/fisiología , Antígenos CD40/metabolismo , Ratones , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/parasitología , Macrófagos/inmunología , Macrófagos/parasitología , Humanos , Femenino , Fosforilación , Interacciones Huésped-Parásitos/inmunología , Sistema de Señalización de MAP Quinasas/inmunología
14.
Plasmid ; 69(1): 90-5, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23159456

RESUMEN

Using the Indian Human immunodeficiency virus type 2 (HIV-2) isolate derived lentiviral vector (LV) system reported earlier, we have derived multiple differently configured transfer vectors. Among the features imparted, the novel ones include a blue/white colony screening platform, a shorter vector backbone candidate and availability of default dual tags. Simultaneously, panels with different utilities were also made using this LV. These include neomycin or puromycin or hygromycin selection markers, with options of default promoter, dual multiple cloning site (MCS) availability and drug inducible transgene expression. All the transfer vectors contain the main MCS with the option of single step sub-cloning of a PCR amplified transgene cassette by T/A cloning strategy apart from cohesive and blunt end cloning sites, as described for the original parent vector. Each transfer vector format was tested by appropriate transgene expression function by transduction of target cells. This is the most comprehensive HIV-2 based lentiviral vector system developed so far and it will significantly aid in preferential applications and thus increase its utility as a versatile system for gene transfer technology.


Asunto(s)
Vectores Genéticos/genética , VIH-2/genética , Lentivirus/genética , Animales , Biomarcadores/metabolismo , Clonación Molecular , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Ratones , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción Genética , Transfección , Transgenes
15.
J Immunol ; 187(3): 1322-32, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21705615

RESUMEN

To reside and multiply successfully within the host macrophages, Leishmania parasites impair the generation of reactive oxygen species (ROS), which are a major host defense mechanism against any invading pathogen. Mitochondrial uncoupling proteins are associated with mitochondrial ROS generation, which is the major contributor of total cellular ROS generation. In the present study we have demonstrated that Leishmania donovani infection is associated with strong upregulation of uncoupling protein 2 (UCP2), a negative regulator of mitochondrial ROS generation located at the inner membrane of mitochondria. Functional knockdown of macrophage UCP2 by small interfering RNA-mediated silencing was associated with increased mitochondrial ROS generation, lower parasite survival, and induction of marked proinflammatory cytokine response. Induction of proinflammatory cytokine response in UCP2 knocked-down cells was a direct consequence of p38 and ERK1/2 MAPK activation, which resulted from ROS-mediated inhibition of protein tyrosine phosphatases (PTPs). Administration of ROS quencher, N-acetyl-l-cysteine, abrogated PTP inhibition in UCP2 knocked-down infected cells, implying a role of ROS in inactivating PTP. Short hairpin RNA-mediated in vivo silencing of UCP2 resulted in decreased Src homology 2 domain-containing tyrosine phosphatase 1 and PTP-1B activity and host-protective proinflammatory cytokine response resulting in effective parasite clearance. To our knowledge, this study, for the first time, reveals the induction of host UCP2 expression during Leishmania infection to downregulate mitochondrial ROS generation, thereby possibly preventing ROS-mediated PTP inactivation to suppress macrophage defense mechanisms.


Asunto(s)
Regulación hacia Abajo/inmunología , Mediadores de Inflamación/fisiología , Canales Iónicos/fisiología , Leishmaniasis Visceral/inmunología , Leishmaniasis Visceral/prevención & control , Mitocondrias/metabolismo , Proteínas Mitocondriales/fisiología , Proteínas Tirosina Fosfatasas/fisiología , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Animales , Línea Celular , Regulación hacia Abajo/genética , Activación Enzimática/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Mediadores de Inflamación/metabolismo , Canales Iónicos/deficiencia , Canales Iónicos/genética , Leishmania donovani/inmunología , Leishmaniasis Visceral/patología , Macrófagos/enzimología , Macrófagos/inmunología , Macrófagos/parasitología , Ratones , Ratones Endogámicos BALB C , Mitocondrias/genética , Mitocondrias/inmunología , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Proteínas Tirosina Fosfatasas/metabolismo , ARN Interferente Pequeño/farmacología , Especies Reactivas de Oxígeno/metabolismo , Proteína Desacopladora 2
16.
mBio ; 14(2): e0016623, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-36976020

RESUMEN

HIV-1 antagonizes SERINC5 by redundant mechanisms, primarily through Nef and additionally via envelope glycoprotein. Paradoxically, HIV-1 preserves Nef function to ensure the exclusion of SERINC5 from virion incorporation regardless of the availability of envelope that can confer resistance, suggesting additional roles of the virion-incorporated host factor. Here, we report an unusual mode of SERINC5 action in inhibiting viral gene expression. This inhibition is observed only in the myeloid lineage cells but not in the cells of epithelial or lymphoid origin. We found that SERINC5-bearing viruses induce the expression of RPL35 and DRAP1 in macrophages, and these host proteins intercept HIV-1 Tat from binding to and recruiting a mammalian capping enzyme (MCE1) to the HIV-1 transcriptional complex. As a result, uncapped viral transcripts are synthesized, leading to the inhibition of viral protein synthesis and subsequent progeny virion biogenesis. Cell-type-specific inhibition of HIV-1 gene expression thus exemplifies a novel antiviral function of virion-incorporated SERINC5. IMPORTANCE In addition to Nef, HIV-1 envelope glycoprotein has been shown to modulate SERINC5-mediated inhibition. Counterintuitively, Nef from the same isolates preserves the ability to prevent SERINC5 incorporation into virions, implying additional functions of the host protein. We identify that virion-associated SERINC5 can manifest an antiviral mechanism independent of the envelope glycoprotein to regulate HIV-1 gene expression in macrophages. This mechanism is exhibited by affecting the viral RNA capping and is plausibly adopted by the host to overcome the envelope glycoprotein-mediated resistance to SERINC5 restriction.


Asunto(s)
Infecciones por VIH , VIH-1 , Animales , Proteínas de la Membrana/metabolismo , VIH-1/fisiología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Antivirales , Macrófagos/metabolismo , Expresión Génica , Glicoproteínas/genética , Mamíferos/metabolismo
17.
Sci Adv ; 9(36): eadh9170, 2023 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-37672576

RESUMEN

The functional consequences of circular RNA (circRNA) expression on HIV-1 replication are largely unknown. Using a customized protocol involving direct RNA nanopore sequencing, here, we captured circRNAs from HIV-1-infected T cells and identified ciTRAN, a circRNA that modulates HIV-1 transcription. We found that HIV-1 infection induces ciTRAN expression in a Vpr-dependent manner and that ciTRAN interacts with SRSF1, a protein known to repress HIV-1 transcription. Our results suggest that HIV-1 hijacks ciTRAN to exclude serine/arginine-rich splicing factor 1 (SRSF1) from the viral transcriptional complex, thereby promoting efficient viral transcription. In addition, we demonstrate that an SRSF1-inspired mimic can inhibit viral transcription regardless of ciTRAN induction. The hijacking of a host circRNA thus represents a previously unknown facet of primate lentiviruses in overcoming transmission bottlenecks.


Asunto(s)
VIH-1 , Nanoporos , Animales , Provirus/genética , VIH-1/genética , ARN Circular , Expresión Génica
18.
Viruses ; 15(3)2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36992361

RESUMEN

The host transmembrane protein SERINC5 is incorporated into retrovirus particles and inhibits HIV-1 infectivity. The lentiviral Nef protein counteracts SERINC5 by downregulating it from the cell surface and preventing its incorporation into virions. The ability of Nef to antagonize the host factor varies in magnitude between different HIV-1 isolates. After having identified a subtype H nef allele unable to promote HIV-1 infectivity in the presence of SERINC5, we investigated the molecular determinants responsible for the defective counteraction of the host factor. Chimeric molecules with a subtype C Nef highly active against SERINC5 were constructed to locate Nef residues crucial for the activity against SERINC5. An Asn at the base of the C-terminal loop of the defective nef allele was found in place of a highly conserved acidic residue (D/E 150). The conversion of Asn to Asp restored the ability of the defective Nef to downregulate SERINC5 and promote HIV-1 infectivity. The substitution was also found to be crucial for the ability of Nef to downregulate CD4, but not for Nef activities that do not rely on the internalization of receptors from the cell surface, suggesting a general implication in promoting clathrin-mediated endocytosis. Accordingly, bimolecular fluorescence complementation revealed that the conserved acidic residue contributes to the recruitment of AP2 by Nef. Altogether, our results confirm that Nef downregulates SERINC5 and CD4 by engaging a similar machinery and indicates that, in addition to the di-leucine motif, other residues in the C-terminal flexible loop are important for the ability of the protein to sustain clathrin-mediated endocytosis.


Asunto(s)
Antígenos CD4 , Linfocitos T CD4-Positivos , VIH-1 , Proteínas de la Membrana , Productos del Gen nef del Virus de la Inmunodeficiencia Humana , Humanos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Proteínas de la Membrana/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/química , Sustitución de Aminoácidos , Células HEK293 , Células Jurkat , VIH-1/patogenicidad , Secuencia de Aminoácidos , Endocitosis , Clatrina , Infecciones por VIH , Antígenos CD4/metabolismo , Regulación hacia Abajo
19.
Mol Ther Nucleic Acids ; 28: 202-218, 2022 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-35402072

RESUMEN

Strategies to modulate cellular DNA repair pathways hold immense potential to enhance the efficiency of CRISPR-Cas9 genome editing platform. In the absence of a repair template, CRISPR-Cas9-induced DNA double-strand breaks are repaired by the endogenous cellular DNA repair pathways to generate loss-of-function edits. Here, we describe a reporter-based assay for expeditious measurement of loss-of-function editing by CRISPR-Cas9. An unbiased chemical screen performed using this assay enabled the identification of small molecules that promote loss-of-function editing. Iterative rounds of screens reveal Repsox, a TGF-ß signaling inhibitor, as a CRISPR-Cas9 editing efficiency enhancer. Repsox invariably increased CRISPR-Cas9 editing in a panel of commonly used cell lines in biomedical research and primary cells. Furthermore, Repsox-mediated editing enhancement in primary human CD4+ T cells enabled the generation of HIV-1-resistant cells with high efficiency. This study demonstrates the potential of transiently targeting cellular pathways by small molecules to improve genome editing for research applications and is expected to benefit gene therapy efforts.

20.
Life Sci Alliance ; 5(7)2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35296517

RESUMEN

Breakthrough infections by emerging SARS-CoV-2 variants raise significant concerns. Here, we sequence-characterized the spike gene from breakthrough infections that corresponded to B.1.617 sublineage. Delineating the functional impact of spike mutations revealed that N-terminal domain (NTD)-specific E156G/Δ157-158 contributed to increased infectivity and reduced sensitivity to vaccine-induced antibodies. A six-nucleotide deletion (467-472) in the spike-coding region introduced this change in the NTD. We confirmed the presence of E156G/Δ157-158 from cases concurrently screened, in addition to other circulating spike (S1) mutations such as T19R, T95I, L452R, E484Q, and D614G. Notably, E156G/Δ157-158 was present in more than 90% of the sequences reported from the USA and UK in October 2021. The spike-pseudotyped viruses bearing a combination of E156G/Δ157-158 and L452R exhibited higher infectivity and reduced sensitivity to neutralization. Notwithstanding, the post-recovery plasma robustly neutralized viral particles bearing the mutant spike. When the spike harbored E156G/Δ157-158 along with L452R and E484Q, increased cell-to-cell fusion was also observed, suggesting a combinatorial effect of these mutations. Our study underscores the importance of non-RBD changes in determining infectivity and immune escape.


Asunto(s)
COVID-19 , Glicoproteína de la Espiga del Coronavirus , Humanos , Mutación , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA