Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Pharm ; 21(2): 831-844, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38174896

RESUMO

Tumor spheroids are promising three-dimensional (3D) in vitro tumor models for the evaluation of drug delivery methods. The design of noninvasive and targeted drug methods is required to improve the intratumoral bioavailability of chemotherapeutic drugs and reduce their adverse off-target effects. Among such methods, microbubble-assisted ultrasound (MB-assisted US) is an innovative modality for noninvasive targeted drug delivery. The aim of the present study is to evaluate the efficacy of this US modality for the delivery of bleomycin, doxorubicin, and irinotecan in colorectal cancer (CRC) spheroids. MB-assisted US permeabilized the CRC spheroids to propidium iodide, which was used as a drug model without affecting their growth and viability. Histological analysis and electron microscopy revealed that MB-assisted US affected only the peripheral layer of the CRC spheroids. The acoustically mediated bleomycin delivery induced a significant decrease in CRC spheroid growth in comparison to spheroids treated with bleomycin alone. However, this US modality did not improve the therapeutic efficacy of doxorubicin and irinotecan on CRC spheroids. In conclusion, this study demonstrates that tumor spheroids are a relevant approach to evaluate the efficacy of MB-assisted US for the delivery of chemotherapeutics.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Irinotecano , Microbolhas , Doxorrubicina/farmacologia , Bleomicina , Esferoides Celulares , Linhagem Celular Tumoral
2.
Cell Mol Life Sci ; 80(12): 353, 2023 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-37940699

RESUMO

The coronavirus' (CoV) membrane (M) protein is the driving force during assembly, but this process remains poorly characterized. Previously, we described two motifs in the C-tail of the Middle East respiratory syndrome CoV (MERS-CoV) M protein involved in its endoplasmic reticulum (ER) exit (211DxE213) and trans-Golgi network (TGN) retention (199KxGxYR204). Here, their function in virus assembly was investigated by two different virus-like particle (VLP) assays and by mutating both motifs in an infectious MERS-CoV cDNA clone. It was shown that the 199KxGxYR204 motif was essential for VLP and infectious virus assembly. Moreover, the mislocalization of the M protein induced by mutation of this motif prevented M-E interaction. Hampering the ER export of M by mutating its 211DxE213 motif still allowed the formation of nucleocapsid-empty VLPs, but prevented the formation of fully assembled VLPs and infectious particles. Taken together, these data show that the MERS-CoV assembly process highly depends on the correct intracellular trafficking of its M protein, and hence that not only specific protein-protein interacting motifs but also correct subcellular localization of the M protein in infected cells is essential for virus formation and should be taken into consideration when studying the assembly process.


Assuntos
Proteínas de Membrana , Coronavírus da Síndrome Respiratória do Oriente Médio , Proteínas de Membrana/metabolismo , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Coronavírus da Síndrome Respiratória do Oriente Médio/metabolismo , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , Proteínas do Nucleocapsídeo/genética , Proteínas do Nucleocapsídeo/metabolismo , Montagem de Vírus/genética
3.
Viruses ; 15(10)2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37896772

RESUMO

IFITMs are a family of highly related interferon-induced transmembrane proteins that interfere with the processes of fusion between viral and cellular membranes and are thus endowed with broad antiviral properties. A number of studies have shown how the antiviral potency of IFITMs is highly dependent on their steady-state levels, their intracellular distribution and a complex pattern of post-translational modifications, parameters that are overall tributary of a number of cellular partners. In an effort to identify additional protein partners involved in the biology of IFITMs, we devised a proteomics-based approach based on the piggyback incorporation of IFITM3 partners into extracellular vesicles. MS analysis of the proteome of vesicles bearing or not bearing IFITM3 identified the NDFIP2 protein adaptor protein as an important regulator of IFITM3 levels. NDFIP2 is a membrane-anchored adaptor protein of the E3 ubiquitin ligases of the NEDD4 family that have already been found to be involved in IFITM3 regulation. We show here that NDFIP2 acts as a recruitment factor for both IFITM3 and NEDD4 and mediates their distribution in lysosomal vesicles. The genetic inactivation and overexpression of NDFIP2 drive, respectively, lower and higher levels of IFITM3 accumulation in the cell, overall suggesting that NDFIP2 locally competes with IFITM3 for NEDD4 binding. Given that NDFIP2 is itself tightly regulated and highly responsive to external cues, our study sheds light on a novel and likely dynamic layer of regulation of IFITM3.


Assuntos
Proteômica , Ubiquitina-Proteína Ligases , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Processamento de Proteína Pós-Traducional , Lisossomos/metabolismo , Antivirais/metabolismo
4.
mBio ; : e0132623, 2023 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-37889017

RESUMO

The human T-lymphotropic virus type 1 (HTLV-1) is an oncogenic retrovirus whose transmission relies primarily on cell-to-cell contacts as cell-free viruses are poorly infectious. Among the intercellular transmission routes described, HTLV-1 biofilms are adhesive structures polarized at the cell surface that confine virions in a protective environment, which is believed to promote their simultaneous delivery during infection. Here, we show that several tetraspanins are enriched in HTLV-1 biofilms and incorporated into the viral envelope. However, we report that only the tetraspanin CD82 interacts with HTLV-1 Gag proteins which initiates their polarization into viral biofilms. Also, we demonstrate that CD82 maintains HTLV-1 biofilm polarization and favors viral transmission, as its silencing induces a complete reorganization of viral clusters at the cell surface and reduces the ability of infected T-cells to transmit the virus. Our results highlight the crucial role of CD82 and its glycosylation state in the architectural organization of HTLV-1 biofilms and their subsequent transfer through intercellular contacts.IMPORTANCEIn the early stages of infection, human T-lymphotropic virus type 1 (HTLV-1) dissemination within its host is believed to rely mostly on cell-to-cell contacts. Past studies unveiled a novel mechanism of HTLV-1 intercellular transmission based on the remodeling of the host-cell extracellular matrix and the generation of cell-surface viral assemblies whose structure, composition, and function resemble bacterial biofilms. These polarized aggregates of infectious virions, identified as viral biofilms, allow the bulk delivery of viruses to target cells and may help to protect virions from immune attacks. However, viral biofilms' molecular and functional description is still in its infancy, although it is crucial to fully decipher retrovirus pathogenesis. Here, we explore the function of cellular tetraspanins (CD9, CD81, CD82) that we detect inside HTLV-1 particles within biofilms. Our results demonstrate specific roles for CD82 in the cell-surface distribution and intercellular transmission of HTLV-1 biofilms, which we document as two essential parameters for efficient viral transmission. At last, our findings indicate that N-glycosylation of cell-surface molecules, including CD82, is required for the polarization of HTLV-1 biofilms and for the efficient transmission of HTLV-1 between T-lymphocytes.

5.
Vet Res ; 54(1): 63, 2023 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-37525204

RESUMO

Chicken infection with Salmonella Typhimurium is an important source of foodborne human diseases. Salmonella colonizes the avian intestinal tract and more particularly the caecum, without causing symptoms. This thus poses a challenge for the prevention of foodborne transmission. Until now, studies on the interaction of Salmonella with the avian gut intestine have been limited by the absence of in vitro intestinal culture models. Here, we established intestinal crypt-derived chicken organoids to better decipher the impact of Salmonella intracellular replication on avian intestinal epithelium. Using a 3D organoid model, we observed a significantly higher replication rate of the intracellular bacteria in caecal organoids than in ileal organoids. Our model thus recreates intracellular environment, allowing Salmonella replication of avian epithelium according to the intestinal segment. Moreover, an inhibition of the cellular proliferation was observed in infected ileal and caecal organoids compared to uninfected organoids. This appears with a higher effect in ileal organoids, as well as a higher cytokine and signaling molecule response in infected ileal organoids at 3 h post-infection (hpi) than in caecal organoids that could explain the lower replication rate of Salmonella observed later at 24 hpi. To conclude, this study demonstrates that the 3D organoid is a model allowing to decipher the intracellular impact of Salmonella on the intestinal epithelium cell response and illustrates the importance of the gut segment used to purify stem cells and derive organoids to specifically study epithelial cell -Salmonella interaction.


Assuntos
Galinhas , Salmonella typhimurium , Humanos , Animais , Salmonella typhimurium/fisiologia , Intestinos , Mucosa Intestinal/microbiologia , Ceco , Organoides/microbiologia
6.
Cell Immunol ; 384: 104658, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36566700

RESUMO

Recent evidence showed that in response to elevated sodium dietary intakes, many body tissues retain Na+ ions for long periods of time and can reach concentrations up to 200 mM. This could modulate the immune system and be responsible for several diseases. However, studies brought contrasted results and the effects of external sodium on human dendritic cell (DC) responses to danger signals remain largely unknown. Considering their central role in triggering T cell response, we tested how NaCl-enriched medium influences human DCs properties. We found that DCs submitted to high extracellular Na+ concentrations up to 200 mM remain viable and maintain the expression of specific DC markers, however, their maturation, chemotaxis toward CCL19, production of pro-inflammatory cytokines and ROS in response to LPS were also partially inhibited. In line with these results, the T-cell allostimulatory capacity of DCs was also inhibited. Finally, our data indicate that high NaCl concentrations triggered the phosphorylation of SGK1 and ERK1/2 kinases. These results raised the possibility that the previously reported pro-inflammatory effects of high NaCl concentrations on T cells might be counterbalanced by a downregulation of DC activation.


Assuntos
Lipopolissacarídeos , Cloreto de Sódio , Humanos , Cloreto de Sódio/farmacologia , Cloreto de Sódio/metabolismo , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/metabolismo , Diferenciação Celular , Cloretos/metabolismo , Cloretos/farmacologia , Células Dendríticas , Citocinas/metabolismo , Sódio/metabolismo , Sódio/farmacologia , Células Cultivadas
7.
Orthop Traumatol Surg Res ; 109(6): 103194, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-34954015

RESUMO

INTRODUCTION: Radial nerve palsy is a classical complication of a humeral shaft fracture. In clinical practice, motor palsy of the radial nerve is sometimes observed without an abnormality felt in the sensory territory. HYPOTHESIS: We hypothesised that this dissociation between sensory and motor involvement is related to anatomical variations of the sensory innervation of the dorsal surface of the first digit space, thus, we decided to study the nature and frequency of these variations. MATERIAL AND METHOD: A cadaveric study was conducted on 24 upper limbs to analyse the truncal origin of the sensory branches innervating the dorsal surface of the first digit space. RESULTS: The sensory branch of the radial nerve (SBRN) participated in the innervation of the dorsal surface of the first digit space in 22 limbs, an anatomical variation was present in 2 cases with a mixed innervation by the SBRN and the lateral cutaneous nerve of forearm (LCNF) in 1 case and singular innervation by LCNF, with no SBRN involvement, in 1 case. Communications between SBRN and LCNF were found in 7 cases. DISCUSSION: Pure motor radial damage, without a sensory deficit of the dorsal surface of the first digit space, does not preclude a complete traumatic injury of the radial nerve. The sensory innervation of this region can be relayed by a branch of the LCNF. LEVEL OF EVIDENCE: IV; cadaveric study.


Assuntos
Antebraço , Neuropatia Radial , Humanos , Antebraço/inervação , Nervo Radial/anatomia & histologia , Nervo Radial/lesões , Polegar , Cadáver
8.
Cell Mol Life Sci ; 79(12): 615, 2022 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-36460928

RESUMO

Although hepatitis E virus (HEV) is the major leading cause of enterically transmitted viral hepatitis worldwide, many gaps remain in the understanding of the HEV lifecycle. Notably, viral factories induced by HEV have not been documented yet, and it is currently unknown whether HEV infection leads to cellular membrane modeling as many positive-strand RNA viruses. HEV genome encodes the ORF1 replicase, the ORF2 capsid protein and the ORF3 protein involved in virion egress. Previously, we demonstrated that HEV produces different ORF2 isoforms including the virion-associated ORF2i form. Here, we generated monoclonal antibodies that specifically recognize the ORF2i form and antibodies that recognize the different ORF2 isoforms. One antibody, named P1H1 and targeting the ORF2i N-terminus, recognized delipidated HEV particles from cell culture and patient sera. Importantly, AlphaFold2 modeling demonstrated that the P1H1 epitope is exposed on HEV particles. Next, antibodies were used to probe viral factories in HEV-producing/infected cells. By confocal microscopy, we identified subcellular nugget-like structures enriched in ORF1, ORF2 and ORF3 proteins and viral RNA. Electron microscopy analyses revealed an unprecedented HEV-induced membrane network containing tubular and vesicular structures. We showed that these structures are dependent on ORF2i capsid protein assembly and ORF3 expression. An extensive colocalization study of viral proteins with subcellular markers, and silencing experiments demonstrated that these structures are derived from the endocytic recycling compartment (ERC) for which Rab11 is a central player. Hence, HEV hijacks the ERC and forms a membrane network of vesicular and tubular structures that might be the hallmark of HEV infection.


Assuntos
Vírus da Hepatite E , Humanos , Vírus da Hepatite E/genética , Compartimentos de Replicação Viral , Proteínas do Capsídeo , Transporte Biológico , Anticorpos Monoclonais
9.
Microbiol Spectr ; 10(5): e0298922, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36040168

RESUMO

Zika virus (ZIKV) is a mosquito-borne flavivirus that emerged in the Pacific islands in 2007 and spread to the Americas in 2015. The infection remains asymptomatic in most cases but can be associated with severe neurological disorders. Despite massive efforts, no specific drug or vaccine against ZIKV infection is available to date. Claudins are tight-junction proteins that favor the entry of several flaviviruses, including ZIKV. In this study, we identified two peptides derived from the N-terminal sequences of claudin-7 and claudin-1, named CL7.1 and CL1.1, respectively, that inhibited ZIKV infection in a panel of human cell lines. Using cell-to-cell fusion assays, we demonstrated that these peptides blocked the ZIKV E-mediated membrane fusion. A comparison of the antiviral efficacy of CL1.1 and CL7.1 pointed to the importance of the peptide amphipathicity. Electron microscopic analysis revealed that CL1.1 altered the ultrastructure of the viral particles likely by binding the virus lipid envelope. However, amphipathicity could not fully explain the antiviral activity of CL1.1. In silico docking simulations suggested that CL1.1 may also interact with the E protein, near its stem region. Overall, our data suggested that claudin-derived peptides inhibition may be linked to simultaneous interaction with the E protein and the viral lipid envelope. Finally, we found that CL1.1 also blocked infection by yellow fever and Japanese encephalitis viruses but not by HIV-1 or SARS-CoV-2. Our results provide a basis for the future development of therapeutics against a wide range of endemic and emerging flaviviruses. IMPORTANCE Zika virus (ZIKV) is a flavivirus transmitted by mosquito bites that have spread to the Pacific Islands and the Americas over the past decade. The infection remains asymptomatic in most cases but can cause severe neurological disorders. ZIKV is a major public health threat in areas of endemicity, and there is currently no specific antiviral drug or vaccine available. We identified two antiviral peptides deriving from the N-terminal sequences of claudin-7 and claudin-1 with the latter being the most effective. These peptides block the envelope-mediated membrane fusion. Our data suggested that the inhibition was likely achieved by simultaneously interacting with the viral lipid envelope and the E protein. The peptides also inhibited other flaviviruses. These results could provide the basis for the development of therapies that might target a wide array of flaviviruses from current epidemics and possibly future emergences.


Assuntos
Claudinas , Fusão de Membrana , Infecção por Zika virus , Zika virus , Humanos , Antivirais/farmacologia , Claudina-1 , Lipídeos , Peptídeos/farmacologia , Infecção por Zika virus/tratamento farmacológico
10.
Virol J ; 19(1): 124, 2022 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-35902969

RESUMO

BACKGROUND: Zika virus (ZIKV), a member of the Flaviviridae family, has caused massive outbreaks of infection in tropical areas over the last decade and has now begun spreading to temperate countries. Little is currently known about the specific host factors involved in the intracellular life cycle of ZIKV. Flaviviridae viruses interact closely with host-cell lipid metabolism and associated secretory pathways. Another Flaviviridae, hepatitis C virus, is highly dependent on apolipoprotein E (ApoE) for the completion of its infectious cycle. We therefore investigated whether ZIKV also interacted with this protein. METHODS: ZIKV infections were performed on both liver and microglia derived cell lines in order to proceed to colocalization analysis and immunoprecipitation assays of ApoE and Zika envelope glycoprotein (Zika E). Transmission electron microscopy combined to immunogold labeling was also performed on the infected cells and related supernatant to study the association of ApoE and Zika E protein in the virus-induced membrane rearrangements and secreted particles, respectively. Finally, the potential of neutralization of anti-ApoE antibodies on ZIKV particles was studied. RESULT: We demonstrated an interaction between ApoE and the Zika E protein. This specific interaction was observed in virus-induced host-cell membrane rearrangements, but also on newly formed intracellular particles. The partial neutralizing effect of anti-ApoE antibody and the immunogold labeling of the two proteins on secreted virions indicates that this interaction is conserved during ZIKV intracellular trafficking and release. CONCLUSIONS: These data suggest that another member of the Flaviviridae also interacts with ApoE, indicating that this could be a common mechanism for the viruses from this family.


Assuntos
Flaviviridae , Infecção por Zika virus , Zika virus , Anticorpos Antivirais , Apolipoproteínas E , Linhagem Celular , Humanos , Proteínas do Envelope Viral , Vírion/metabolismo
11.
Cell Mol Life Sci ; 79(8): 425, 2022 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-35841484

RESUMO

Positive single-strand RNA (+ RNA) viruses can remodel host cell membranes to induce a replication organelle (RO) isolating the replication of their genome from innate immunity mechanisms. Some of these viruses, including severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), induce double-membrane vesicles (DMVs) for this purpose. Viral non-structural proteins are essential for DMV biogenesis, but they cannot form without an original membrane from a host cell organelle and a significant supply of lipids. The endoplasmic reticulum (ER) and the initial mechanisms of autophagic processes have been shown to be essential for the biogenesis of SARS-CoV-2 DMVs. However, by analogy with other DMV-inducing viruses, it seems likely that the Golgi apparatus, mitochondria and lipid droplets are also involved. As for hepatitis C virus (HCV), pores crossing both membranes of SARS-CoV-2-induced DMVs have been identified. These pores presumably allow the supply of metabolites essential for viral replication within the DMV, together with the export of the newly synthesized viral RNA to form the genome of future virions. It remains unknown whether, as for HCV, DMVs with open pores can coexist with the fully sealed DMVs required for the storage of large amounts of viral RNA. Interestingly, recent studies have revealed many similarities in the mechanisms of DMV biogenesis and morphology between these two phylogenetically distant viruses. An understanding of the mechanisms of DMV formation and their role in the infectious cycle of SARS-CoV-2 may be essential for the development of new antiviral approaches against this pathogen or other coronaviruses that may emerge in the future.


Assuntos
COVID-19 , Hepatite C , Retículo Endoplasmático/metabolismo , Hepacivirus/genética , Humanos , RNA Viral/genética , RNA Viral/metabolismo , SARS-CoV-2 , Proteínas não Estruturais Virais/genética , Replicação Viral
12.
Nature ; 603(7903): 900-906, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35296858

RESUMO

Infections of the central nervous system are among the most serious infections1,2, but the mechanisms by which pathogens access the brain remain poorly understood. The model microorganism Listeria monocytogenes (Lm) is a major foodborne pathogen that causes neurolisteriosis, one of the deadliest infections of the central nervous system3,4. Although immunosuppression is a well-established host risk factor for neurolisteriosis3,5, little is known about the bacterial factors that underlie the neuroinvasion of Lm. Here we develop a clinically relevant experimental model of neurolisteriosis, using hypervirulent neuroinvasive strains6 inoculated in a humanized mouse model of infection7, and we show that the bacterial surface protein InlB protects infected monocytes from Fas-mediated cell death by CD8+ T cells in a manner that depends on c-Met, PI3 kinase and FLIP. This blockade of specific anti-Lm cellular immune killing lengthens the lifespan of infected monocytes, and thereby favours the transfer of Lm from infected monocytes to the brain. The intracellular niche that is created by InlB-mediated cell-autonomous immune resistance also promotes Lm faecal shedding, which accounts for the selection of InlB as a core virulence gene of Lm. We have uncovered a specific mechanism by which a bacterial pathogen confers an increased lifespan to the cells it infects by rendering them resistant to cell-mediated immunity. This promotes the persistence of Lm within the host, its dissemination to the central nervous system and its transmission.


Assuntos
Doenças do Sistema Nervoso Central , Listeria monocytogenes , Listeriose , Animais , Proteínas de Bactérias/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Doenças do Sistema Nervoso Central/microbiologia , Modelos Animais de Doenças , Listeria monocytogenes/patogenicidade , Listeriose/microbiologia , Camundongos , Monócitos , Virulência
13.
J Biol Chem ; 298(1): 101500, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34929171

RESUMO

In HIV, the polyprotein precursor Gag orchestrates the formation of the viral capsid. In the current view of this viral assembly, Gag forms low-order oligomers that bind to the viral genomic RNA triggering the formation of high-ordered ribonucleoprotein complexes. However, this assembly model was established using biochemical or imaging methods that do not describe the cellular location hosting Gag-gRNA complex nor distinguish gRNA packaging in single particles. Here, we studied the intracellular localization of these complexes by electron microscopy and monitored the distances between the two partners by morphometric analysis of gold beads specifically labeling Gag and gRNA. We found that formation of these viral clusters occurred shortly after the nuclear export of the gRNA. During their transport to the plasma membrane, the distance between Gag and gRNA decreases together with an increase of gRNA packaging. Point mutations in the zinc finger patterns of the nucleocapsid domain of Gag caused an increase in the distance between Gag and gRNA as well as a sharp decrease of gRNA packaged into virions. Finally, we show that removal of stem loop 1 of the 5'-untranslated region does not interfere with gRNA packaging, whereas combined with the removal of stem loop 3 is sufficient to decrease but not abolish Gag-gRNA cluster formation and gRNA packaging. In conclusion, this morphometric analysis of Gag-gRNA cluster formation sheds new light on HIV-1 assembly that can be used to describe at nanoscale resolution other viral assembly steps involving RNA or protein-protein interactions.


Assuntos
Produtos do Gene gag , HIV-1 , Nucleoproteínas , Regiões 5' não Traduzidas , Produtos do Gene gag/genética , Produtos do Gene gag/metabolismo , Genômica , HIV-1/genética , HIV-1/metabolismo , Microscopia Eletrônica de Transmissão , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , RNA Guia de Cinetoplastídeos , RNA Viral/genética , RNA Viral/metabolismo , Montagem de Vírus/genética
14.
Curr Opin Microbiol ; 66: 11-20, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34923331

RESUMO

Listeria monocytogenes (Lm) is a foodborne pathogen and the etiological agent of listeriosis. This facultative intracellular Gram-positive bacterium has the ability to colonize the intestinal lumen, cross the intestinal, blood-brain and placental barriers, leading to bacteremia, neurolisteriosis and maternal-fetal listeriosis. Lm is a model microorganism for the study of the interplay between a pathogenic microbe, host tissues and microbiota in vivo. Here we review how animal models permissive to Lm-host interactions allow deciphering some of the key steps of the infectious process, from the intestinal lumen to the crossing of host barriers and dissemination within the host. We also highlight recent investigations using tagged Lm and clinically relevant strains that have shed light on within-host dynamics and the purifying selection of Lm virulence factors. Studying Lm infection in vivo is a way forward to explore host biology and unveil the mechanisms that have selected its capacity to closely associate with its vertebrate hosts.


Assuntos
Listeria monocytogenes , Listeriose , Animais , Proteínas de Bactérias , Feminino , Interações Hospedeiro-Patógeno , Listeria monocytogenes/genética , Placenta , Gravidez
15.
Sci Rep ; 11(1): 21856, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34750487

RESUMO

Hepatitis C is a major threat to public health for which an effective treatment is available, but a prophylactic vaccine is still needed to control this disease. We designed a vaccine based on chimeric HBV-HCV envelope proteins forming subviral particles (SVPs) that induce neutralizing antibodies against HCV in vitro. Here, we aimed to increase the neutralizing potential of those antibodies, by using HBV-HCV SVPs bearing apolipoprotein E (apoE). These particles were produced by cultured stable mammalian cell clones, purified and characterized. We found that apoE was able to interact with both chimeric HBV-HCV (E1-S and E2-S) proteins, and with the wild-type HBV S protein. ApoE was also detected on the surface of purified SVPs and improved the folding of HCV envelope proteins, but its presence lowered the incorporation of E2-S protein. Immunization of New Zealand rabbits resulted in similar anti-S responses for all rabbits, whereas anti-E1/-E2 antibody titers varied according to the presence or absence of apoE. Regarding the neutralizing potential of these anti-E1/-E2 antibodies, it was higher in rabbits immunized with apoE-bearing particles. In conclusion, the association of apoE with HCV envelope proteins may be a good strategy for improving HCV vaccines based on viral envelope proteins.


Assuntos
Apolipoproteínas E/administração & dosagem , Apolipoproteínas E/imunologia , Hepacivirus/imunologia , Vírus da Hepatite B/imunologia , Vacinas contra Hepatite Viral/administração & dosagem , Vacinas contra Hepatite Viral/imunologia , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Neutralizantes/sangue , Apresentação de Antígeno/imunologia , Linhagem Celular , Feminino , Hepatite C/imunologia , Hepatite C/prevenção & controle , Anticorpos Anti-Hepatite C/biossíntese , Anticorpos Anti-Hepatite C/sangue , Humanos , Evasão da Resposta Imune , Coelhos , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/imunologia , Proteínas do Envelope Viral/administração & dosagem , Proteínas do Envelope Viral/imunologia
16.
Orthop Traumatol Surg Res ; 107(8): 103074, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34563733

RESUMO

INTRODUCTION: The unique anatomical characteristics of the forearm bones makes their reconstruction challenging. The aim of this study was to report the surgical methods and results of the induced membrane technique applied to traumatic forearm bone defects. MATERIAL AND METHODS: We evaluated retrospectively a case series of 13 patients operated between 2010 and 2017. The first surgical step consisted of debridement of the fracture site and implantation of a cement spacer with appropriate fixation. The anatomy of the forearm skeleton had to be restored. The second step, done 6 weeks later, consisted of removing the cement spacer and applying cancellous bone autograft harvested from the iliac crest. The outcome measures were radiological bone union, need for surgical revision, and postoperative wrist range of motion. RESULTS: All 13 patients were men, with a mean age of 39 years (18-67). The average follow-up was 2.5 years. Eleven patients were suffering from a nonunion and two from a post-traumatic bone defect. Six patients had an identified preoperative infection. Three patients had previously undergone an unsuccessful treatment for their nonunion with bone addition. The maximum length of bone reconstruction was 12 cm. Union was achieved in 12 of 13 patients in a mean of 5 months (3-8). The other patient died during the postoperative course. Two patients needed revision surgery: ulnar shortening osteotomy (1 case) and additional tendon reconstruction (1 case). The mean pronosupination range was 123° on average (55-180°). The mean flexion-extension range was 106° (90-130°). CONCLUSION: The induced membrane technique is a reliable reconstruction technique that is well suited to reconstruction of the forearm skeleton. LEVEL OF EVIDENCE: IV, retrospective study.


Assuntos
Antebraço , Fraturas não Consolidadas , Adulto , Transplante Ósseo/métodos , Fraturas não Consolidadas/cirurgia , Humanos , Masculino , Amplitude de Movimento Articular , Estudos Retrospectivos , Resultado do Tratamento , Articulação do Punho
17.
J Biol Chem ; 297(3): 101081, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34403699

RESUMO

The human APOBEC3A (A3A) cytidine deaminase is a powerful DNA mutator enzyme recognized as a major source of somatic mutations in tumor cell genomes. However, there is a discrepancy between APOBEC3A mRNA levels after interferon stimulation in myeloid cells and A3A detection at the protein level. To understand this difference, we investigated the expression of two novel alternative "A3Alt" proteins encoded in the +1-shifted reading frame of the APOBEC3A gene. A3Alt-L and its shorter isoform A3Alt-S appear to be transmembrane proteins targeted to the mitochondrial compartment that induce membrane depolarization and apoptosis. Thus, the APOBEC3A gene represents a new example wherein a single gene encodes two proapoptotic proteins, A3A cytidine deaminases that target the genome and A3Alt proteins that target mitochondria.


Assuntos
Citidina Desaminase/genética , Citidina Desaminase/fisiologia , Mitocôndrias/genética , Proteínas/genética , Proteínas/fisiologia , Apoptose/genética , Proteínas Reguladoras de Apoptose/genética , Citidina Desaminase/metabolismo , DNA/genética , Mutação da Fase de Leitura/genética , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Genoma/genética , Humanos , Mitocôndrias/metabolismo , Mutação/genética , Proteínas/metabolismo , RNA Mensageiro/genética , Fases de Leitura/genética
18.
Microorganisms ; 9(6)2021 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-34063935

RESUMO

CC17 Streptococcus agalactiae carrying group-A prophages is increasingly responsible for neonatal infections. To investigate the impact of the genetic features of a group-A prophage, we first conducted an in silico analysis of the genome of 12/111phiA, a group-A prophage carried by a strain responsible for a bloodstream infection in a parturient. This revealed a Restriction Modification system, suggesting a prophage maintenance strategy and five ORFs of interest for the host and encoding a type II toxin antitoxin system RelB/YafQ, an endonuclease, an S-adenosylmethionine synthetase MetK, and an StrP-like adhesin. Using the WT strain cured from 12/111phiA and constructing deleted mutants for the ORFs of interest, and their complemented mutants, we demonstrated an impact of prophage features on growth characteristics, cell morphology and biofilm formation. Our findings argue in favor of 12/111phiA domestication by the host and a role of prophage features in cell autoaggregation, glycocalyx and biofilm formation. We suggest that lysogeny may promote GBS adaptation to the acid environment of the vagina, consequently colonizing and infecting neonates.

19.
BMC Microbiol ; 21(1): 153, 2021 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-34020586

RESUMO

BACKGROUND: Salmonella can invade host cells via a type three secretion system called T3SS-1 and its outer membrane proteins, PagN and Rck. However, the mechanism of PagN-dependent invasion pathway used by Salmonella enterica, subspecies enterica serovar Typhimurium remains unclear. RESULTS: Here, we report that PagN is well conserved and widely distributed among the different species and subspecies of Salmonella. We showed that PagN of S. Typhimurium was sufficient and necessary to enable non-invasive E. coli over-expressing PagN and PagN-coated beads to bind to and invade different non-phagocytic cells. According to the literature, PagN is likely to interact with heparan sulfate proteoglycan (HSPG) as PagN-mediated invasion could be inhibited by heparin treatment in a dose-dependent manner. This report shows that this interaction is not sufficient to allow the internalization mechanism. Investigation of the role of ß1 integrin as co-receptor showed that mouse embryo fibroblasts genetically deficient in ß1 integrin were less permissive to PagN-mediated internalization. Moreover, PagN-mediated internalization was fully inhibited in glycosylation-deficient pgsA-745 cells treated with anti-ß1 integrin antibody, supporting the hypothesis that ß1 integrin and HSPG cooperate to induce the PagN-mediated internalization mechanism. In addition, use of specific inhibitors and expression of dominant-negative derivatives demonstrated that tyrosine phosphorylation and class I phosphatidylinositol 3-kinase were crucial to trigger PagN-dependent internalization, as for the Rck internalization mechanism. Finally, scanning electron microscopy with infected cells showed microvillus-like extensions characteristic of Zipper-like structure, engulfing PagN-coated beads and E. coli expressing PagN, as observed during Rck-mediated internalization. CONCLUSIONS: Our results supply new comprehensions into T3SS-1-independent invasion mechanisms of S. Typhimurium and highly indicate that PagN induces a phosphatidylinositol 3-kinase signaling pathway, leading to a Zipper-like entry mechanism as the Salmonella outer membrane protein Rck.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Infecções por Salmonella/microbiologia , Salmonella typhimurium/metabolismo , Animais , Proteínas da Membrana Bacteriana Externa/genética , Linhagem Celular , Fibroblastos/metabolismo , Fibroblastos/microbiologia , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Camundongos , Infecções por Salmonella/genética , Infecções por Salmonella/metabolismo , Salmonella typhimurium/genética , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/metabolismo
20.
C R Biol ; 343(4): 79-89, 2021 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-33988325

RESUMO

Chikungunya is an infectious disease caused by the chikungunya virus (CHIKV), an alphavirus transmitted to humans by Aedes mosquitoes, and for which there is no licensed vaccine nor antiviral treatments. By using a loss-of-function genetic screen, we have recently identified the FHL1 protein as an essential host factor for CHIKV tropism and pathogenesis. FHL1 is highly expressed in muscles cells and fibroblasts, the main CHIKV-target cells. FHL1 interacts with the viral protein nsP3 and plays a critical role in CHIKV genome amplification. Experiments in vivo performed in FHL1-deficient mice have shown that these animals are resistant to infection and do not develop muscular lesions. Altogether these observations, published in the journal Nature [1], show that FHL1 is a key host factor for CHIKV pathogenesis and identify the interaction between FHL1 and nsP3 as a promising target for the development of new antiviral strategies.


Le chikungunya est une maladie infectieuse causée par le virus chikungunya (CHIKV), un alphavirus transmis à l'Homme par les moustiques Aedes et contre lequel il n'existe ni vaccin, ni traitements antiviraux. En utilisant une approche de crible génétique par perte de fonction, nous avons récemment identifié la protéine FHL1 comme un facteur cellulaire essentiel pour le tropisme et la pathogénèse du CHIKV. FHL1 est une molécule présente majoritairement dans les cellules musculaires et les fibroblastes, les cibles privilégiées de CHIKV. FHL1 interagit avec la protéine virale nsP3 et joue un rôle décisif dans le mécanisme d'amplification du génome de CHIKV. Des expériences in vivo chez des souris déficientes pour FHL1 ont montré que ces animaux sont résistants à l'infection et ne développent pas de lésions musculaires. L'ensemble de ces observations publiées dans la revue Nature [1] montrent que FHL1 est un facteur cellulaire clé pour la pathogénèse de CHIKV et identifient l'interaction entre FHL1 et nsp3 comme une cible prometteuse pour le développement de nouvelles stratégies antivirales.


Assuntos
Febre de Chikungunya , Vírus Chikungunya , Animais , Vírus Chikungunya/genética , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas com Domínio LIM , Camundongos , Proteínas Musculares , Tropismo , Proteínas não Estruturais Virais , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA