Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 57
Filtrer
1.
bioRxiv ; 2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38979149

RÉSUMÉ

The early stages of HIV-1 infection include the trafficking of the viral core into the nucleus of infected cells. However, much remains to be understood about how HIV-1 accomplishes nuclear import and the consequences of the import pathways utilized on nuclear events. The host factor cleavage and polyadenylation specificity factor 6 (CPSF6) assists HIV-1 nuclear localization and post-entry integration targeting. Here, we used a CPSF6 truncation mutant lacking a functional nuclear localization signal (NLS), CPSF6-358, and appended heterologous NLSs to rescue nuclear localization. We show that some, but not all, NLSs drive CPSF6-358 into the nucleus. Interestingly, we found that some nuclear localized CPSF6-NLS chimeras supported inefficient HIV-1 infection. We found that HIV-1 still enters the nucleus in these cell lines but fails to traffic to speckle-associated domains (SPADs). Additionally, we show that HIV-1 fails to efficiently integrate in these cell lines. Collectively, our results demonstrate that the NLS of CPSF6 facilitates steps of HIV-1 infection subsequent to nuclear import and additionally identify the ability of canonical NLS sequences to influence cargo localization in the nucleus following nuclear import.

2.
Methods Mol Biol ; 2807: 141-151, 2024.
Article de Anglais | MEDLINE | ID: mdl-38743226

RÉSUMÉ

To integrate with host chromatin and establish a productive infection, HIV-1 must translocate the viral Ribonucleoprotein (RNP) complex through the nuclear pore complex (NPC). Current assay to measure HIV-1 nuclear import relies on a transient byproduct of HIV-1 integration failure called 2-LTR circles. However, 2-LTR circles require complete or near-complete reverse transcription and association with the non-homologous end joining (NHEJ) machinery in the nucleus, which can complicate interpretation of 2-LTR circle formation as a measure of nuclear import kinetics. Here, we describe an approach to measure nuclear import of infectious HIV-1 particles. This involves chemically induced dimerization of Nup62, a central FG containing nucleoporin. Using this technique, nuclear import of infectious particles can be monitored in both primary and cell culture models. In response to host factor depletion or restriction factors, changes in HIV-1 nuclear import can be effectively measured using the nuclear import kinetics (NIK) assay.


Sujet(s)
Transport nucléaire actif , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Complexe protéique du pore nucléaire , Pore nucléaire , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Humains , Pore nucléaire/métabolisme , Complexe protéique du pore nucléaire/métabolisme , Cinétique , Noyau de la cellule/métabolisme , Infections à VIH/virologie , Infections à VIH/métabolisme , Intégration virale
3.
Inflamm Res ; 73(4): 563-580, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38411635

RÉSUMÉ

BACKGROUND: Altered expression of vacuole membrane protein 1 (VMP1) has recently been observed in the context of multiple sclerosis and Parkinson's disease (PD). However, how changes in VMP1 expression may impact pathogenesis has not been explored. OBJECTIVE: This study aimed to characterize how altered VMP1 expression affects NLRP3 inflammasome activation and mitochondrial function. METHODS: VMP1 expression was depleted in a monocytic cell line using CRISPR-Cas9. The effect of VMP1 on NLRP3 inflammasome activation was examined by stimulating cells with LPS and ATP or α-synuclein fibrils. Inflammasome activation was determined by caspase-1 activation using both a FLICA assay and a biosensor as well as by the release of proinflammatory molecules measured by ELISA. RNA-sequencing was utilized to define global gene expression changes resulting from VMP1 deletion. SERCA activity and mitochondrial function were investigated using various fluorescence microscopy-based approaches including a novel method that assesses the function of individual mitochondria in a cell. RESULTS: Here, we report that genetic deletion of VMP1 from a monocytic cell line resulted in increased NLRP3 inflammasome activation and release of proinflammatory molecules. Examination of the VMP1-dependent changes in these cells revealed that VMP1 deficiency led to decreased SERCA activity and increased intracellular [Ca2+]. We also observed calcium overload in mitochondria in VMP1 depleted cells, which was associated with mitochondrial dysfunction and release of mitochondrial DNA into the cytoplasm and the extracellular environment. CONCLUSIONS: Collectively, these studies reveal VMP1 as a negative regulator of inflammatory responses, and we postulate that decreased expression of VMP1 can aggravate the inflammatory sequelae associated with neurodegenerative diseases like PD.


Sujet(s)
Inflammasomes , Maladies mitochondriales , Humains , Inflammasomes/métabolisme , Protéines membranaires/métabolisme , Maladies mitochondriales/génétique , Maladies mitochondriales/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Vacuoles/métabolisme
4.
bioRxiv ; 2023 Dec 02.
Article de Anglais | MEDLINE | ID: mdl-37693451

RÉSUMÉ

Viruses exploit host cytoskeletal elements and motor proteins for trafficking through the dense cytoplasm. Yet the molecular mechanism that describes how viruses connect to the motor machinery is unknown. Here, we demonstrate the first example of viral microtubule trafficking from purified components: HIV-1 hijacking microtubule transport machinery. We discover that HIV-1 directly binds to the retrograde microtubule-associated motor, dynein, and not via a cargo adaptor, as previously suggested. Moreover, we show that HIV-1 motility is supported by multiple, diverse dynein cargo adaptors as HIV-1 binds to dynein light and intermediate chains on dynein's tail. Further, we demonstrate that multiple dynein motors tethered to rigid cargoes, like HIV-1 capsids, display reduced motility, distinct from the behavior of multiple motors on membranous cargoes. Our results introduce a new model of viral trafficking wherein a pathogen opportunistically 'hijacks' the microtubule transport machinery for motility, enabling multiple transport pathways through the host cytoplasm.

5.
Cell Death Dis ; 14(9): 632, 2023 09 25.
Article de Anglais | MEDLINE | ID: mdl-37749127

RÉSUMÉ

A severe complication of hematopoietic stem cell transplantation is graft-versus-host disease (GvHD), a reaction that occurs following the transfer of donor immune cells (the graft) into an allogeneic host. Transplanted cells recognize host alloantigens as foreign, resulting in the activation of donor T cells and migration of these pathological cells into host tissues. In this study, we found that caspase-1 is activated in alloreactive murine and human CD4+ and CD8+ T cells early during acute GvHD (aGvHD). The presence of inflammasome-bound active caspase-1 (p33) and ASC-speck formation confirmed inflammasome activation in these cells. We further measured gasdermin D (GSDMD) cleavage and IL-18 secretion from alloreactive T cells ex vivo. Isolated T cells with high levels of active caspase-1 had a strong inflammatory transcriptional signature and a metabolic phenotype similar to inflammatory myeloid cells, including the upregulation of proinflammatory cytokines and metabolic switch from oxidative phosphorylation to aerobic glycolysis. We also observed oxidative stress, mitochondrial dysfunction, and cell death phenotypes consistent with inflammatory cell death in alloreactive T cells. For the first time, this study characterizes caspase-1 activation in transplanted T cells during aGvHD, using mouse and human models, adding to a body of literature supporting inflammasome function in cells of the adaptive immune system.


Sujet(s)
Lymphocytes T CD8+ , Maladie du greffon contre l'hôte , Humains , Animaux , Souris , Inflammasomes , Caspase-1 , Lymphocytes T CD4+
6.
J Neuroinflammation ; 20(1): 196, 2023 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-37635235

RÉSUMÉ

BACKGROUND: Individuals who have experienced mild traumatic brain injuries (mTBIs) suffer from several comorbidities, including chronic pain. Despite extensive studies investigating the underlying mechanisms of mTBI-associated chronic pain, the role of inflammation in long-term pain after mTBIs is not fully elucidated. Given the shifting dynamics of inflammation, it is important to understand the spatial-longitudinal changes in inflammatory processes following mTBIs and their effects on TBI-related pain. METHODS: We utilized a recently developed transgenic caspase-1 luciferase reporter mouse model to monitor caspase-1 activation through a thinned skull window in the in vivo setting following three closed-head mTBI events. Organotypic coronal brain slice cultures and acutely dissociated dorsal root ganglion (DRG) cells provided tissue-relevant context of inflammation signal. Mechanical allodynia was assessed by mechanical withdrawal threshold to von Frey and thermal hyperalgesia withdrawal latency to radiant heat. Mouse grimace scale (MGS) was used to detect spontaneous or non-evoked pain. In some experiments, mice were prophylactically treated with MCC950, a potent small molecule inhibitor of NLRP3 inflammasome assembly to inhibit injury-induced inflammatory signaling. Bioluminescence spatiotemporal dynamics were quantified in the head and hind paws, and caspase-1 activation was confirmed by immunoblot. Immunofluorescence staining was used to monitor the progression of astrogliosis and microglial activation in ex vivo brain tissue following repetitive closed-head mTBIs. RESULTS: Mice with repetitive closed-head mTBIs exhibited significant increases of the bioluminescence signals within the brain and paws in vivo for at least one week after each injury. Consistently, immunoblotting and immunofluorescence experiments confirmed that mTBIs led to caspase-1 activation, astrogliosis, and microgliosis. Persistent changes in MGS and hind paw withdrawal thresholds, indicative of pain states, were observed post-injury in the same mTBI animals in vivo. We also observed enhanced inflammatory responses in ex vivo brain slice preparations and DRG for at least 3 days following mTBIs. In vivo treatment with MCC950 significantly reduced caspase-1 activation-associated bioluminescent signals in vivo and decreased stimulus-evoked and non-stimulus evoked nociception. CONCLUSIONS: Our findings suggest that the inflammatory states in the brain and peripheral nervous system following repeated mTBIs are coincidental with the development of nociceptive sensitization, and that these events can be significantly reduced by inhibition of NLRP3 inflammasome activation.


Sujet(s)
Commotion de l'encéphale , Lésions traumatiques de l'encéphale , Douleur chronique , Animaux , Souris , Gliose , Inflammasomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Nociception , Hyperalgésie/étiologie , Caspase-1
7.
Res Sq ; 2023 Jan 27.
Article de Anglais | MEDLINE | ID: mdl-36747822

RÉSUMÉ

Altered expression of vacuole membrane protein 1 (VMP1) has recently been observed in the context of multiple sclerosis and Parkinson's disease (PD). However, how changes in VMP1 expression may impact pathogenesis has not been explored. Here, we report that genetic deletion of VMP1 from a monocytic cell line resulted in increased NLRP3 inflammasome activation and release of proinflammatory molecules. Examination of the VMP1 dependent changes in these cells revealed that VMP1 deficiency led to decreased SERCA activity and increased intracellular [Ca2+]. We also observed calcium overload in mitochondria in VMP1 depleted cells, which was associated with mitochondrial dysfunction and release of mitochondrial DNA into the cytoplasm and the extracellular environment. Autophagic defects were also observed in VMP1 depleted macrophages. Collectively, these studies reveal VMP1 as a negative regulator of inflammatory responses, and we postulate that decreased expression of VMP1 can aggravate the inflammatory sequelae associated with neurodegenerative diseases like PD.

8.
Curr Opin Virol ; 53: 101203, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-35121335

RÉSUMÉ

A hallmark feature of lentiviruses, which separates them from other members of the retrovirus family, is their ability to infect non-dividing cells by traversing the nuclear pore complex. The viral determinant that mediates HIV-1 nuclear import is the viral capsid (CA) protein, which forms the conical core protecting the HIV-1 genome in a mature virion. Recently, a series of novel approaches developed to monitor post-fusion events in infection have challenged previous textbook models of the viral life cycle, which envisage reverse transcription and disassembly of the capsid core as events that complete in the cytoplasm. In this review, we summarize these recent findings and describe their implications on our understanding of the spatiotemporal staging of HIV-1 infection with a focus on the nuclear import and its implications in other aspects of the viral lifecycle.


Sujet(s)
Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Transport nucléaire actif , Capside/métabolisme , Protéines de capside/génétique , Noyau de la cellule , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Humains , Pore nucléaire/métabolisme , Transcription inverse
9.
Autophagy ; 18(5): 1020-1048, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-34612142

RÉSUMÉ

Numerous lines of evidence support the premise that the misfolding and subsequent accumulation of SNCA/α-synuclein (synuclein alpha) is responsible for the underlying neuronal pathology observed in Parkinson disease (PD) and other synucleinopathies. Moreover, the cell-to-cell transfer of these misfolded SNCA species is thought to be responsible for disease progression and the spread of cellular pathology throughout the brain. Previous work has shown that when exogenous, misfolded SNCA fibrils enter cells through endocytosis, they can damage and rupture the membranes of their endocytotic vesicles in which they are trafficked. Rupture of these vesicular membranes exposes intralumenal glycans leading to galectin protein binding, subsequent autophagic protein recruitment, and, ultimately, their introduction into the autophagic-lysosomal pathway. Increasing evidence indicates that both pathological and non-pathological SNCA species undergo autophagy-dependent unconventional secretion. While other proteins have also been shown to be secreted from cells by autophagy, what triggers this release process and how these specific proteins are recruited to a secretory autophagic pathway is largely unknown. Here, we use a human midbrain dopamine (mDA) neuronal culture model to provide evidence in support of a cellular mechanism that explains the cell-to-cell transfer of pathological forms of SNCA that are observed in PD. We demonstrate that LGALS3 (galectin 3) mediates the release of SNCA following vesicular damage. SNCA release is also dependent on TRIM16 (tripartite motif containing 16) and ATG16L1 (autophagy related 16 like 1), providing evidence that secretion of SNCA is mediated by an autophagic secretory pathway.


Sujet(s)
Neurones dopaminergiques , Galectine -3 , Maladie de Parkinson , alpha-Synucléine , Autophagie/physiologie , Protéines du sang , Neurones dopaminergiques/métabolisme , Galectine -3/métabolisme , Galectines , Humains , Lysosomes/métabolisme , Mésencéphale/métabolisme , Maladie de Parkinson/métabolisme , Protéines à motif tripartite/métabolisme , Ubiquitin-protein ligases/métabolisme , alpha-Synucléine/métabolisme
10.
J Neuroinflammation ; 18(1): 263, 2021 Nov 10.
Article de Anglais | MEDLINE | ID: mdl-34758843

RÉSUMÉ

BACKGROUND: Inflammatory bowel disease (IBD) is established to drive pathological sequelae in organ systems outside the intestine, including the central nervous system (CNS). Many patients exhibit cognitive deficits, particularly during disease flare. The connection between colonic inflammation and neuroinflammation remains unclear and characterization of the neuroinflammatory phenotype in the brain during colitis is ill-defined. METHODS: Transgenic mice expressing a bioluminescent reporter of active caspase-1 were treated with 2% dextran sodium sulfate (DSS) for 7 days to induce acute colitis, and colonic, systemic and neuroinflammation were assessed. In some experiments, mice were prophylactically treated with paquinimod (ABR-215757) to inhibit S100A9 inflammatory signaling. As a positive control for peripheral-induced neuroinflammation, mice were injected with lipopolysaccharide (LPS). Colonic, systemic and brain inflammatory cytokines and chemokines were measured by cytokine bead array (CBA) and Proteome profiler mouse cytokine array. Bioluminescence was quantified in the brain and caspase activation was confirmed by immunoblot. Immune cell infiltration into the CNS was measured by flow cytometry, while light sheet microscopy was used to monitor changes in resident microglia localization in intact brains during DSS or LPS-induced neuroinflammation. RNA sequencing was performed to identify transcriptomic changes occurring in the CNS of DSS-treated mice. Expression of inflammatory biomarkers were quantified in the brain and serum by qRT-PCR, ELISA and WB. RESULTS: DSS-treated mice exhibited clinical hallmarks of colitis, including weight loss, colonic shortening and inflammation in the colon. We also detected a significant increase in inflammatory cytokines in the serum and brain, as well as caspase and microglia activation in the brain of mice with ongoing colitis. RNA sequencing of brains isolated from DSS-treated mice revealed differential expression of genes involved in the regulation of inflammatory responses. This inflammatory phenotype was similar to the signature detected in LPS-treated mice, albeit less robust and transient, as inflammatory gene expression returned to baseline following cessation of DSS. Pharmacological inhibition of S100A9, one of the transcripts identified by RNA sequencing, attenuated colitis severity and systemic and neuroinflammation. CONCLUSIONS: Our findings suggest that local inflammation in the colon drives systemic inflammation and neuroinflammation, and this can be ameliorated by inhibition of the S100 alarmin, S100A9.


Sujet(s)
Encéphale/physiopathologie , Calgranuline B/génétique , Colite/induit chimiquement , Colite/prévention et contrôle , Maladies neuro-inflammatoires/prévention et contrôle , Maladies neuro-inflammatoires/physiopathologie , Quinoléines/usage thérapeutique , Animaux , Marqueurs biologiques , Caspase-1/métabolisme , Chimiokines/métabolisme , Colite/physiopathologie , Cytokines/métabolisme , Sulfate dextran , Humains , Lipopolysaccharides , Souris , Souris de lignée C57BL , Souris transgéniques
11.
J Immunol ; 206(9): 2101-2108, 2021 05 01.
Article de Anglais | MEDLINE | ID: mdl-33846225

RÉSUMÉ

Acute graft-versus-host disease (aGvHD) is a severe, often lethal, complication of hematopoietic stem cell transplantation, and although prophylactic regimens are given as standard pretransplantation therapy, up to 60% of these patients develop aGvHD, and require additional immunosuppressive intervention. We treated mice with a purified probiotic molecule, exopolysaccharide (EPS) from Bacillus subtilis, shortly before and after induction of aGvHD and found that, whereas only 10% of control mice survived to day 80, 70% of EPS-treated mice survived to 80 d. EPS treatment of donor-only mice resulted in ∼60% survival. Using a biosensor mouse model to assess inflammation in live mice during aGvHD, we found that EPS prevented the activation of alloreactive donor T cells. In vitro, EPS did not affect T cells directly but, instead, induced bone marrow-derived dendritic cells (BMDCs) that displayed characteristics of inhibitory dendritic cells (DCs). Development of these BMDCs required TLR4 signaling through both MyD88 and TRIF pathways. Using BMDCs derived from IDO knockout mice, we showed that T cell inhibition by EPS-treated BMDCs was mediated through the suppressive effects of IDO. These studies describe a bacterial molecule that modulates immune responses by inducing inhibitory DCs in a TLR4-dependent manner, and these cells have the capacity to inhibit T cell activation through IDO. We suggest that EPS or EPS-treated DCs can serve as novel agents for preventing aGvHD.


Sujet(s)
Bacillus subtilis/composition chimique , Maladie du greffon contre l'hôte/immunologie , Polyosides bactériens/immunologie , Animaux , Bacillus subtilis/immunologie , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris knockout
12.
J Extracell Vesicles ; 9(1): 1789326, 2020 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-32944176

RÉSUMÉ

Extracellular vesicles (EVs) have been implicated in a wide variety of biological activities, have been implicated in the pathogenesis of numerous diseases, and have been proposed to serve as potential biomarkers of disease in human patients and animal models. However, characterization of EV populations is often performed using methods that do not account for the heterogeneity of EV populations and require comparatively large sample sizes to facilitate analysis. Here, we describe an imaging-based method that allows for the multiplexed characterization of EV populations at the single EV level following centrifugation of EV populations directly onto cover slips, allowing comprehensive analysis of EV populations with relatively small samples. We observe that canonical EV markers are present on subsets of EVs which differ substantially in a producer cell and cargo specific fashion, including differences in EVs containing different HIV-1 proteins previously reported to be incorporated into pathogenic EVs. We also describe a lectin binding assay to interrogate EVs based on their glycan content, which we observe to change in response to pharmacological modulation of secretory autophagy pathways. These studies collectively reveal that a multiplexed analysis of EV populations using fluorescent microscopy can reveal differences in specific EV populations that may be used to understand the biogenesis of specific EV populations and/or to interrogate small subsets of EVs of interest within larger EV populations in biological samples.

13.
Nat Microbiol ; 5(9): 1088-1095, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32483230

RÉSUMÉ

Retroviral infection involves the reverse transcription of the viral RNA genome into DNA, which is subsequently integrated into the host cell genome. Human immunodeficiency virus type 1 (HIV-1) and other lentiviruses mediate the infection of non-dividing cells through the ability of the capsid protein1 to engage the cellular nuclear import pathways of the target cell and mediate their nuclear translocation through components of the nuclear pore complex2-4. Although recent studies have observed the presence of the capsid protein in the nucleus during infection5-8, reverse transcription and disassembly of the viral core have conventionally been considered to be cytoplasmic events. Here, we use an inducible nuclear pore complex blockade to monitor the kinetics of HIV-1 nuclear import and define the biochemical staging of these steps of infection. Surprisingly, we observe that nuclear import occurs with relatively rapid kinetics (<5 h) and precedes the completion of reverse transcription in target cells, demonstrating that reverse transcription is completed in the nucleus. We also observe that HIV-1 remains susceptible to the capsid-destabilizing compound PF74 following nuclear import, revealing that uncoating is completed in the nucleus. Additionally, we observe that certain capsid mutants are insensitive to a Nup62-mediated nuclear pore complex blockade in cells that potently block infection by wild-type capsid, demonstrating that HIV-1 can use distinct nuclear import pathways during infection. These studies collectively define the spatio-temporal staging of critical steps of HIV-1 infection and provide an experimental system to separate and thereby define the cytoplasmic and nuclear stages of infection by other viruses.


Sujet(s)
Noyau de la cellule/métabolisme , Infections à VIH/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Pore nucléaire/métabolisme , Pore nucléaire/virologie , Transcription inverse , Transport nucléaire actif , Lymphocytes T CD4+/virologie , Capside/métabolisme , Protéines de capside/métabolisme , Cytoplasme/métabolisme , Cellules HEK293 , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Cellules HeLa , Interactions hôte-pathogène , Humains , Indoles , Macrophages/virologie , Phénylalanine/analogues et dérivés , Réplication virale
14.
Cell Rep ; 31(12): 107810, 2020 06 23.
Article de Anglais | MEDLINE | ID: mdl-32579936

RÉSUMÉ

Cellular metabolism governs the susceptibility of CD4 T cells to HIV-1 infection. Multiple early post-fusion steps of HIV-1 replication are restricted in resting peripheral blood CD4 T cells; however, molecular mechanisms that underlie metabolic control of these steps remain undefined. Here, we show that mTOR activity following T cell stimulatory signals overcomes metabolic restrictions in these cells by enabling the expansion of dNTPs to fuel HIV-1 reverse transcription (RT), as well as increasing acetyl-CoA to stabilize microtubules that transport RT products. We find that catalytic mTOR inhibition diminishes the expansion of pools of both of these metabolites by limiting glucose and glutamine utilization in several pathways, thereby suppressing HIV-1 infection. We demonstrate how mTOR-coordinated biosyntheses enable the early steps of HIV-1 replication, add metabolic mechanisms by which mTOR inhibitors block HIV-1, and identify some metabolic modules downstream of mTOR as druggable targets for HIV-1 inhibition.


Sujet(s)
VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Espace intracellulaire/métabolisme , Transcription inverse/génétique , Sérine-thréonine kinases TOR/métabolisme , Acétyl coenzyme A/métabolisme , Acétylation , Transport biologique , Lymphocytes T CD4+/virologie , Cytokines/métabolisme , Rétrocontrôle physiologique , Glycolyse , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Humains , Lysine/métabolisme , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Complexe-2 cible mécanistique de la rapamycine/métabolisme , Microtubules/métabolisme , Modèles biologiques , Récepteurs aux antigènes des cellules T/métabolisme , Tubuline/métabolisme , Réplication virale/génétique
15.
AIDS Res Hum Retroviruses ; 36(7): 545-549, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32414291

RÉSUMÉ

One cannot spend >5 min on social media at the moment without finding a link to some conspiracy theory or other regarding the origin of SARS-CoV2, the coronavirus responsible for the COVID-19 pandemic. From the virus being deliberately released as a bioweapon to pharmaceutical companies blocking the trials of natural remedies to boost their dangerous drugs and vaccines, the Internet is rife with far-fetched rumors. And predictably, now that the first immunization trials have started, the antivaccine lobby has latched on to most of them. In the last week, the trailer for a new "bombshell documentary" Plandemic has been doing the rounds, gaining notoriety for being repeatedly removed from YouTube and Facebook. We usually would not pay much heed to such things, but for retrovirologists like us, the name associated with these claims is unfortunately too familiar: Dr. Judy Mikovits.


Sujet(s)
Betacoronavirus , Infections à coronavirus/épidémiologie , Syndrome de fatigue chronique/virologie , Escroquerie , Personnel de laboratoire d'analyses médicales/psychologie , Pneumopathie virale/épidémiologie , Tumeurs de la prostate/virologie , Personnel de recherche/psychologie , Infections à Retroviridae/complications , Retroviridae/physiologie , Animaux , COVID-19 , Infections à coronavirus/virologie , Tromperie , Humains , Mâle , Souris , Pandémies , Pneumopathie virale/virologie , Infections à Retroviridae/virologie , SARS-CoV-2 , Médias sociaux
16.
Nat Microbiol ; 4(12): 2044-2051, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-31636416

RÉSUMÉ

The HIV-1 capsid (CA) protein lattice encases viral genomic RNA and regulates steps essential to target-cell invasion1. Cyclophilin A (CypA) has interacted with the CA of lentiviruses related to HIV-1 for millions of years2-7. Disruption of the CA-CypA interaction decreases HIV-1 infectivity in human cells8-12 but stimulates infectivity in non-human primate cells13-15. Genetic and biochemical data suggest that CypA protects HIV-1 from a CA-specific restriction factor in human cells16-20. Discovery of the CA-specific restriction factor tripartite-containing motif 5α (TRIM5α)21 and multiple, independently derived, TRIM5-CypA fusion genes4,5,15,22-26 pointed to human TRIM5α being the CypA-sensitive restriction factor. However, HIV-1 restriction by human TRIM5α in tumour cell lines is minimal21 and inhibition of such activity by CypA has not been detected27. Here, by exploiting reverse genetic tools optimized for primary human blood cells, we demonstrate that disruption of the CA-CypA interaction renders HIV-1 susceptible to potent restriction by human TRIM5α, with the block occurring before reverse transcription. Endogenous TRIM5α associated with virion cores as they entered the cytoplasm, but only when the CA-CypA interaction was disrupted. These experiments resolve the long-standing mystery of the role of CypA in HIV-1 replication by demonstrating that this ubiquitous cellular protein shields HIV-1 from previously inapparent restriction by human TRIM5α.


Sujet(s)
Protéines de capside/métabolisme , Cyclophiline A/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Protéines à motif tripartite/métabolisme , Ubiquitin-protein ligases/métabolisme , Réplication virale , Facteurs de restriction antiviraux , Protéines de capside/génétique , Cellules cultivées , Cyclophiline A/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Cellules HeLa , Humains , Macrophages/virologie , ARN viral/génétique , Génétique inverse , Transcription inverse , Protéines à motif tripartite/génétique , Ubiquitin-protein ligases/génétique , Virion
17.
J Immunol ; 203(9): 2497-2507, 2019 11 01.
Article de Anglais | MEDLINE | ID: mdl-31562211

RÉSUMÉ

Inflammasomes are multiprotein complexes that coordinate cellular inflammatory responses and mediate host defense. Following recognition of pathogens and danger signals, inflammasomes assemble and recruit and activate caspase-1, the cysteine protease that cleaves numerous downstream targets, including pro-IL-1ß and pro-IL-18 into their biologically active form. In this study, we sought to develop a biosensor that would allow us to monitor the initiation, progression, and resolution of inflammation in living animals. To this end, we inserted a known caspase-1 target sequence into a circularly permuted luciferase construct that becomes bioluminescent upon protease cleavage. This biosensor was activated in response to various inflammatory stimuli in human monocytic cell lines and murine bone marrow-derived macrophages. Next, we generated C57BL/6 transgenic mice constitutively expressing the caspase-1 biosensor. We were able to monitor the spatiotemporal dynamics of caspase-1 activation and onset of inflammation in individual animals in the context of a systemic bacterial infection, colitis, and acute graft-versus-host disease. These data established a model whereby the development and progression of inflammatory responses can be monitored in the context of these and other mouse models of disease.


Sujet(s)
Techniques de biocapteur/méthodes , Caspase-1/analyse , Inflammation/étiologie , Animaux , Apoptose , Colite/enzymologie , Évolution de la maladie , Maladie du greffon contre l'hôte/enzymologie , Cellules HEK293 , Humains , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Infections à staphylocoques/enzymologie , Cellules THP-1
18.
J Virol ; 93(14)2019 07 15.
Article de Anglais | MEDLINE | ID: mdl-31068426

RÉSUMÉ

TRIM5α is an antiviral restriction factor that inhibits retroviral infection in a species-specific fashion. TRIM5α binds to and forms assemblies around the retroviral capsid. Following binding, poorly understood, ubiquitin-dependent events lead to the disassembly of the viral core, prior to the accumulation of viral reverse transcription products in the target cell. It is also known that assemblies of TRIM5α and other TRIM family proteins can be targets of autophagic degradation. The goal of this study was to define the role of specific ubiquitin linkages in the retroviral restriction and autophagic degradation of TRIM5α and delineate any connection between these two processes. To this end, we generated fusion proteins in which the catalytic domains of different deubiquitinase (DUB) enzymes, with different specificities for polyubiquitinated linkages, were fused to the N-terminal RING domain of Rhesus macaque TRIM5α. We assessed the role of ubiquitination in restriction and the degree to which specific types of ubiquitination are required for the association of TRIM5α with autophagic proteins. We determined that K63-linked ubiquitination by TRIM5α is required to induce capsid disassembly and to inhibit reverse transcription of HIV, while the ability to inhibit HIV-1 infection was not dependent on K63-linked ubiquitination. We also observed that K63-linked ubiquitination is required for the association of TRIM5α with autophagosomal membranes and the autophagic adapter protein p62.IMPORTANCE Although the mechanisms by which TRIM5α can induce the abortive disassembly of retroviral capsids have remained obscure, numerous studies have suggested a role for ubiquitination and cellular degradative pathways. These studies have typically relied on global perturbation of cellular degradative pathways. Here, through the use of linkage-specific deubiquitinating enzymes tethered to TRIM5α, we delineate the ubiquitin linkages which drive specific steps in restriction and degradation by TRIM5α, providing evidence for a noncanonical role for K63-linked ubiquitin in the process of retroviral restriction by TRIM5α and potentially providing insight into the mechanism of action of other TRIM family proteins.


Sujet(s)
Capside/métabolisme , Infections à VIH/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Polyubiquitine/métabolisme , Transcription inverse , Protéines à motif tripartite/métabolisme , Ubiquitin-protein ligases/métabolisme , Cellules A549 , Animaux , Autophagosomes/métabolisme , Autophagosomes/virologie , Cellules HEK293 , Infections à VIH/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Cellules HeLa , Humains , Macaca mulatta , Polyubiquitine/génétique , Protéines à motif tripartite/génétique , Ubiquitin-protein ligases/génétique , Ubiquitination/génétique
19.
PLoS One ; 14(3): e0212888, 2019.
Article de Anglais | MEDLINE | ID: mdl-30889178

RÉSUMÉ

TRIM5α is an interferon inducible restriction factor which contributes to intrinsic defense against HIV infection by targeting the HIV capsid protein CA. Although human TRIM5α (huTRIM5α) does not potently inhibit HIV-1 infection, the ability of huTRIM5α to exhibit some control of HIV-1 infection is evidenced by a single nucleotide polymorphism in huTRIM5α which substitutes aspartic acid to glycine at position 249 (G249D) in the L2 region and is associated with higher susceptibility to HIV-1 infection. To understand the mechanistic basis for the reduced antiviral activity, we employed biophysical and cell biological methods coupled with molecular dynamics simulations to compare WT and the G249D polymorphism of huTRIM5α. We investigated the differences in conformational dynamics of rhesus and huTRIM5α Coiled Coil-Linker 2 (CC-L2) dimers utilizing circular dichroism and single molecule-Fluorescence Energy Transfer (sm-FRET). These methods revealed that the G249D dimer exhibits secondary structure and conformational dynamics similar to WT huTRIM5α. Homology modelling revealed that G249 was present on the hairpin of the antiparallel dimer, in a position which may act to stabilize the adjacent BBox2 domain which mediates the inter-dimeric contacts required for the formation of TRIM5 assemblies. We therefore asked if the G249D mutant forms assemblies in cells with the same efficiency as WT protein by expressing these proteins as YFP fusions and quantifying the number of assemblies in cells. In cells expressing comparable amounts of protein, the G249D mutant formed fewer assemblies than WT protein, in agreement with our homology modeling predictions and molecular dynamics simulations of dimers and higher oligomers of TRIM5α, providing a mechanistic explanation of the reduced antiviral activity of the G249D polymorphism.


Sujet(s)
Protéines de transport/génétique , Infections à VIH/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Animaux , Facteurs de restriction antiviraux , Protéines de capside/immunologie , Protéines de capside/métabolisme , Protéines de transport/immunologie , Protéines de transport/métabolisme , Chats , Prédisposition génétique à une maladie , Cellules HEK293 , Infections à VIH/immunologie , Infections à VIH/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Protéines du virus de l'immunodéficience humaine/immunologie , Protéines du virus de l'immunodéficience humaine/métabolisme , Humains , Simulation de dynamique moléculaire , Polymorphisme de nucléotide simple , Structure en hélice alpha/génétique , Domaines protéiques/génétique , Structure quaternaire des protéines/génétique , Protéines recombinantes/génétique , Protéines recombinantes/immunologie , Protéines recombinantes/métabolisme , Protéines à motif tripartite , Ubiquitin-protein ligases
20.
J Virol ; 92(16)2018 08 15.
Article de Anglais | MEDLINE | ID: mdl-29899089

RÉSUMÉ

Recent studies show that human immunodeficiency virus type 1 (HIV-1) can utilize microtubules and their associated proteins to complete key postfusion steps during infection. These include associating with both dynein and kinesin motors, as well as proteins, which enhance infection by altering microtubule dynamics during infection. In this article, we will discuss findings on how dynein and kinesin motors, as well as other microtubule-associated proteins, influence HIV-1 trafficking, viral core uncoating, and nuclear import of the viral ribonucleoprotein (RNP).


Sujet(s)
Infections à VIH/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Interactions hôte-pathogène , Protéines associées aux microtubules/métabolisme , Microtubules/métabolisme , Transport nucléaire actif , Humains , Protéines virales , Décapsidation virale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE