Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Am J Respir Cell Mol Biol ; 66(4): 439-451, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35081328

RESUMEN

Persistent neutrophilic inflammation associated with chronic pulmonary infection causes progressive lung injury and, eventually, death in individuals with cystic fibrosis (CF), a genetic disease caused by biallelic mutations in the CF transmembrane conductance regulator (CFTR) gene. Therefore, we examined whether roscovitine, a cyclin-dependent kinase inhibitor that (in other conditions) reduces inflammation while promoting host defense, might provide a beneficial effect in the context of CF. Herein, using CFTR-depleted zebrafish larvae as an innovative vertebrate model of CF immunopathophysiology, combined with murine and human approaches, we sought to determine the effects of roscovitine on innate immune responses to tissue injury and pathogens in the CF condition. We show that roscovitine exerts antiinflammatory and proresolution effects in neutrophilic inflammation induced by infection or tail amputation in zebrafish. Roscovitine reduces overactive epithelial reactive oxygen species (ROS)-mediated neutrophil trafficking by reducing DUOX2/NADPH-oxidase activity and accelerates inflammation resolution by inducing neutrophil apoptosis and reverse migration. It is important to note that, although roscovitine efficiently enhances intracellular bacterial killing of Mycobacterium abscessus in human CF macrophages ex vivo, we found that treatment with roscovitine results in worse infection in mouse and zebrafish models. By interfering with DUOX2/NADPH oxidase-dependent ROS production, roscovitine reduces the number of neutrophils at infection sites and, consequently, compromises granuloma formation and maintenance, favoring extracellular multiplication of M. abscessus and more severe infection. Our findings bring important new understanding of the immune-targeted action of roscovitine and have significant therapeutic implications for safely targeting inflammation in CF.


Asunto(s)
Infecciones por Mycobacterium no Tuberculosas , Neutrófilos , Animales , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Oxidasas Duales , Ratones , Infecciones por Mycobacterium no Tuberculosas/microbiología , Roscovitina/farmacología , Roscovitina/uso terapéutico , Pez Cebra
2.
Dev Dyn ; 250(5): 701-716, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33369805

RESUMEN

BACKGROUND: In zebrafish, lymphatic endothelial cells (LECs) originate from multiple/several distinct progenitor populations and generate organ-specific lymphatic vasculatures. Cell fate and tissue specificities were determined using a combination of genetically engineered transgenic lines in which the promoter of a LEC-specific gene drives expression of a fluorescent reporter protein. RESULTS: We established a novel zebrafish transgenic line expressing eGFP under the control of part of the zebrafish batf3 promoter (Basic Leucine Zipper ATF-Like Transcription Factor 3). Spatiotemporal examination of Tg(batf3MIN:eGFP) transgenic fish revealed a typical lymphatic expression pattern, which does not perfectly recapitulate the expression pattern of existing LEC transgenic lines. eGFP+ cells constitute a heterogeneous endothelial cell population, which expressed LEC and/or blood endothelial cells (BEC) markers in different tissues. In addition, we characterize the renal eGFP+ cell as a population of interest to study kidney diseases and regeneration. CONCLUSION: Our Tg(batf3MIN:eGFP) reporter zebrafish line provides a useful system to study LEC populations, of which heterogeneity depends on origin of progenitors, tissue environment and physiological conditions. We further developed a novel fish-adapted tissue clearing method, which allows deep imaging and 3D-visualization of vascular and lymphatic networks in the whole organism.


Asunto(s)
Células Endoteliales , Genes Reporteros , Vasos Linfáticos/citología , Pez Cebra , Animales , Animales Modificados Genéticamente
3.
J Neurochem ; 155(2): 137-153, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-31811775

RESUMEN

The olfactory mucosa, where the first step of odor detection occurs, is a privileged pathway for environmental toxicants and pathogens toward the central nervous system. Indeed, some pathogens can infect olfactory sensory neurons including their axons projecting to the olfactory bulb allowing them to bypass the blood-brain barrier and reach the central nervous system (CNS) through the so-called olfactory pathway. The respiratory syncytial virus (RSV) is a major respiratory tract pathogen but there is growing evidence that RSV may lead to CNS impairments. However, the mechanisms involved in RSV entering into the CNS have been poorly described. In this study, we wanted to explore the capacity of RSV to reach the CNS via the olfactory pathway and to better characterize RSV cellular tropism in the nasal cavity. We first explored the distribution of RSV infectious sites in the nasal cavity by in vivo bioluminescence imaging and a tissue clearing protocol combined with deep-tissue imaging and 3D image analyses. This whole tissue characterization was confirmed with immunohistochemistry and molecular biology approaches. Together, our results provide a novel 3D atlas of mouse nasal cavity anatomy and show that RSV can infect olfactory sensory neurons giving access to the central nervous system by entering the olfactory bulb. Cover Image for this issue: doi: 10.1111/jnc.14765.


Asunto(s)
Mucosa Olfatoria/inervación , Mucosa Olfatoria/virología , Neuronas Receptoras Olfatorias/virología , Virus Sincitiales Respiratorios , Animales , Sistema Nervioso Central/diagnóstico por imagen , Sistema Nervioso Central/virología , Enfermedades del Sistema Nervioso Central/diagnóstico por imagen , Enfermedades del Sistema Nervioso Central/virología , Femenino , Cabeza/anatomía & histología , Imagenología Tridimensional , Ratones , Ratones Endogámicos BALB C , Mucosa Nasal/virología , Bulbo Olfatorio/virología , Mucosa Olfatoria/diagnóstico por imagen , ARN Viral/aislamiento & purificación , Tropismo , Replicación Viral
4.
Fish Shellfish Immunol ; 86: 724-733, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30550990

RESUMEN

Tripartite motif (TRIM) family or RBCC proteins comprises characteristic zinc-binding domains (a RING (R), a B-box type 1 (B1) and a B-box type 2 (B2)) and coiled-coil (CC) domain followed by a C-terminus variable domain. There are about 80 different TRIM proteins in human, but more than 200 in zebrafish with several large gene expansions (ftr >70 genes; btr >30 genes; trim35 > 30 genes). Repertoires of trim genes in fish are variable across fishes, but they have been remarkably diversified independently in a number of species. In mammals, TRIM proteins are involved in antiviral immunity through an astonishing diversity of mechanisms, from direct viral restriction to modulation of immune signaling and more recently autophagy. In fish, the antiviral role of TRIM proteins remains poorly understood. In zebrafish, fish specific TRIMs so called fintrims show a signature of positive selection in the C terminus SPRY domain, reminding features of mammalian antiviral trims such as TRIM5. Expression studies show that a number of trim genes, including many fintrims, can be induced during viral infections, and may play a role in antiviral defence. Some of them trigger antiviral activity in vitro against DNA and RNA viruses, such as FTR83 that also up-regulates the expression of type I IFN in zebrafish larvae. The tissue distribution of TRIM expression suggests that they may be involved in the regionalization of antiviral immunity, providing a particular protection to sensitive areas exposed to invading pathogens.


Asunto(s)
Evolución Molecular , Proteínas de Peces/genética , Peces/genética , Proteínas de Motivos Tripartitos/genética , Animales , Antivirales/metabolismo , Proteínas de Peces/metabolismo , Peces/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
5.
BMC Cancer ; 18(1): 1237, 2018 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-30526524

RESUMEN

BACKGROUND: The current treatment of malignant melanoma is limited by the lack of effective therapeutic approaches, and alternative treatments are needed. Proliferative diseases such as melanoma and other cancers may be treatable by virally-encoded apoptotic proteins that are targeted to rapidly multiplying cells. Caspase-dependent apoptosis, that is frequently used in chemotherapy, can boost the cell proliferation that caspase-independent cell death does not. METHODS: In the current study, the porcine circovirus type 2 (PCV2), proapoptotic protein ORF3 was expressed in mouse and human cancer cell lines, and its apoptotic activity was assessed. RESULTS: Quantitative assessment of the apoptotic cells by flow cytometry showed that apoptotic cell death was significantly increased in ORF3-expressing malignant cells, compared to ORF3 non-expressing cells. Our data show that PCV2 ORF3 induces apoptosis in a caspase-3 and -8 independent manner. ORF3 expression seems to cause an increase in abnormal mitosis in B16F10 melanoma cells by interacting with centrosomes and thereby disrupting the formation of the mitotic spindle. In addition, we show that ORF3 of PCV2 also exhibits significant anti-tumor effects in vivo. Although the expression of Regulator of G protein Signaling (RGS)-16 by recipient mice inhibited the development of grafted melanoma in vivo, it was not required for the antitumoral activity of ORF3. CONCLUSION: PCV2 ORF3 causes abnormal mitosis in rapidly dividing cells and increases the apoptosis of cancer cells. Apoptin might, therefore, be considered to develop future antitumoral strategies.


Asunto(s)
Apoptosis , Circovirus , Melanoma/patología , Proteínas Virales , Animales , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Viroterapia Oncolítica/métodos , Virus Oncolíticos
6.
J Immunol ; 192(9): 4328-41, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24683187

RESUMEN

Ease of imaging and abundance of genetic tools make the zebrafish an attractive model host to understand host-pathogen interactions. However, basic knowledge regarding the identity of genes involved in antiviral immune responses is still lagging in this species. We conducted a microarray analysis of the larval zebrafish response to two models of RNA virus infections with very different outcomes. Chikungunya virus (CHIKV) induces a rapid and protective IFN response. Infection with infectious hematopoietic necrosis virus is lethal and is associated with a delayed and inefficient IFN response. A typical signature of IFN-stimulated genes (ISGs) was observed with both viruses, but was stronger for CHIKV. We further compared the zebrafish and human ISG repertoires and made a genomic and phylogenic characterization of the main gene families. We describe a core set of well-induced ISGs conserved across vertebrates, as well as multigenic families diversified independently in each taxon. The conservation of ISGs involved in antiviral signaling indicates conservation of the main feedback loops in these pathways. Whole-mount in situ hybridization of selected transcripts in infected larvae revealed a typical pattern of expression for ISGs in the liver, gut, and blood vessels with both viruses. We further show that some inflammatory genes were additionally induced through IFN-independent pathways by infectious hematopoietic necrosis virus and not by CHIKV. This study provides a useful reference set for the analysis of host-virus interactions in zebrafish and highlights the differences between protective and nonprotective antiviral innate responses.


Asunto(s)
Infecciones por Alphavirus/genética , Inmunidad Innata/genética , Interferones/genética , Infecciones por Rhabdoviridae/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Infecciones por Alphavirus/inmunología , Animales , Fiebre Chikungunya , Regulación de la Expresión Génica , Humanos , Inmunidad Innata/inmunología , Hibridación in Situ , Virus de la Necrosis Hematopoyética Infecciosa/inmunología , Interferones/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Filogenia , Reacción en Cadena en Tiempo Real de la Polimerasa , Infecciones por Rhabdoviridae/inmunología , Pez Cebra/inmunología , Pez Cebra/virología , Proteínas de Pez Cebra/inmunología
7.
Virologie (Montrouge) ; 20(6): 321-334, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-32187968

RESUMEN

Real time analysis of viral infection is now feasible with the emergence of viral mutants encoding reporter genes (such as fluorescent proteins or Luciferase). The development of reverse genetic approaches in the virology field has contributed significantly to improve our knowledge on viral dissemination in mammals using intravital imaging technologies. In this context, zebra fish recently appeared as a promizing tool to study infectiology and viruses responsible for human diseases such as Herpes, Flu, hepatitis and chikungunya infections. This small vertebrate is optically transparent during the early stages of development, and has been particularly useful to study the tropism of fluorescent viruses, spreading mechanisms as well as viral persistence at cellular resolution. The size of this small organism is compatible with imaging of the whole larva using a dissecting microscope. Genome editing technology has contributed important tools that are now being exploited for the characterization of the host pathogen interaction in vivo, mainly focused on the conserved features of antiviral innate immunity.

8.
J Gen Virol ; 94(Pt 6): 1435-1440, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23388201

RESUMEN

In naturally acquired transmissible spongiform encephalopathies, the pathogenic agents or prions spread from the sites of initial peripheral uptake or replication to the brain where they cause progressive and fatal neurodegeneration. Routing via the peripheral nervous system is considered to be one of the main pathways to the central nervous system. Replication of prions in Schwann cells is viewed as a potentially important mechanism for efficient prion spread along nerves. Here we used a Cre-loxP mouse transgenetic approach to disrupt host-encoded prion protein (PrP(C)) specifically in myelinating Schwann cells. Despite the use of infection routes targeting highly myelinated nerves, there was no alteration in mouse prion pathogenesis, suggesting that conversion-dependent, centripetal spread of prions does not crucially rely on PrP(C) expressed by myelinating Schwann cells.


Asunto(s)
Marcación de Gen , Vaina de Mielina/metabolismo , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Enfermedades por Prión/metabolismo , Enfermedades por Prión/patología , Células de Schwann/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Transgénicos , Vaina de Mielina/patología
9.
Virologie (Montrouge) ; 17(6): 426-441, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-31910607

RESUMEN

The rainbow trout (Oncorhynchus mykiss) is one of most significant model in fish immunology, as well as a key species in European aquaculture. Two well-known Novirhabdoviruses, the viral haemorrhagic septicemia virus (VHSV) and the infectious hematopoietic necrosis virus (IHNV) cause serious damage in fish farms and represent a significant threat for aquaculture in a number of countries. A significant variability of resistance to these infections - intra- and inter-populations - has been described in rainbow trout. This genetic variability is not only relevant for the selection of fish naturally resistant to these viruses, but also for the dissection of the mechanisms of fish antiviral immunity. The methods used to analyze the genetic bases of the trout resistance to VHSV, and primary results are reported here from the identification of a major QTL to the impact of type I interferons.

10.
Dis Model Mech ; 16(5)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37264878

RESUMEN

Hereditary hemorrhagic telangiectasia (HHT) is a rare genetic disease caused by mutations affecting components of bone morphogenetic protein (BMP)/transforming growth factor-ß (TGF-ß) signaling in endothelial cells. This disorder is characterized by arteriovenous malformations that are prone to rupture, and the ensuing hemorrhages are responsible for iron-deficiency anemia. Along with activin receptor-like kinase (ALK1), mutations in endoglin are associated with the vast majority of HHT cases. In this study, we characterized the zebrafish endoglin locus and demonstrated that it produces two phylogenetically conserved protein isoforms. Functional analysis of a CRISPR/Cas9 zebrafish endoglin mutant revealed that Endoglin deficiency is lethal during the course from juvenile stage to adulthood. Endoglin-deficient zebrafish develop cardiomegaly, resulting in heart failure and hypochromic anemia, which both stem from chronic hypoxia. endoglin mutant zebrafish display structural alterations of the developing gills and underlying vascular network that coincide with hypoxia. Finally, phenylhydrazine treatment demonstrated that lowering hematocrit/blood viscosity alleviates heart failure and enhances the survival of Endoglin-deficient fish. Overall, our data link Endoglin deficiency to heart failure and establish zebrafish as a valuable HHT model.


Asunto(s)
Insuficiencia Cardíaca , Telangiectasia Hemorrágica Hereditaria , Animales , Endoglina/genética , Endoglina/metabolismo , Telangiectasia Hemorrágica Hereditaria/complicaciones , Telangiectasia Hemorrágica Hereditaria/genética , Pez Cebra , Células Endoteliales/metabolismo , Insuficiencia Cardíaca/metabolismo , Receptores de Activinas Tipo II/genética
11.
J Virol ; 85(7): 3077-85, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21248032

RESUMEN

Infection by prions involves conversion of a host-encoded cell surface protein (PrP(C)) to a disease-related isoform (PrP(Sc)). PrP(C) carries two glycosylation sites variably occupied by complex N-glycans, which have been suggested by previous studies to influence the susceptibility to these diseases and to determine characteristics of prion strains. We used the Rov cell system, which is susceptible to sheep prions, to generate a series of PrP(C) glycosylation mutants with mutations at one or both attachment sites. We examined their subcellular trafficking and ability to convert into PrP(Sc) and to sustain stable prion propagation in the absence of wild-type PrP. The susceptibility to infection of mutants monoglycosylated at either site differed dramatically depending on the amino acid substitution. Aglycosylated double mutants showed overaccumulation in the Golgi compartment and failed to be infected. Introduction of an ectopic glycosylation site near the N terminus fully restored cell surface expression of PrP but not convertibility into PrP(Sc), while PrP(C) with three glycosylation sites conferred cell permissiveness to infection similarly to the wild type. In contrast, predominantly aglycosylated molecules with nonmutated N-glycosylation sequons, produced in cells expressing glycosylphosphatidylinositol-anchorless PrP(C), were able to form infectious PrP(Sc). Together our findings suggest that glycosylation is important for efficient trafficking of anchored PrP to the cell surface and sustained prion propagation. However, properly trafficked glycosylation mutants were not necessarily prone to conversion, thus making it difficult in such studies to discern whether the amino acid changes or glycan chain removal most influences the permissiveness to prion infection.


Asunto(s)
Mutación Missense , Priones/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Línea Celular , Membrana Celular/química , Glicosilación , Aparato de Golgi/química , Priones/genética , Transporte de Proteínas , Conejos , Ovinos
12.
ACS Nano ; 16(12): 20470-20487, 2022 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-36459488

RESUMEN

Cargo transport by molecular motors along microtubules is essential for the function of eukaryotic cells, in particular neurons in which axonal transport defects constitute the early pathological features of neurodegenerative diseases. Mainly studied in motor and sensory neurons, axonal transport is still difficult to characterize in neurons of the brain in absence of appropriate in vivo tools. Here, we measured fast axonal transport by tracing the second harmonic generation (SHG) signal of potassium titanyl phosphate (KTP) nanocrystals (nanoKTP) endocytosed by brain neurons of zebrafish (Zf) larvae. Thanks to the optical translucency of Zf larvae and to the perfect photostability of nanoKTP SHG, we achieved a high scanning speed of 20 frames (of ≈90 µm × 60 µm size) per second in Zf brain. We focused our study on endolysosomal vesicle transport in axons of known polarization, separately analyzing kinesin and dynein motor-driven displacements. To validate our assay, we used either loss-of-function mutations of dynein or kinesin 1 or the dynein inhibitor dynapyrazole and quantified several transport parameters. We successfully demonstrated that dynapyrazole reduces the nanoKTP mobile fraction and retrograde run length consistently, while the retrograde run length increased in kinesin 1 mutants. Taking advantage of nanoKTP SHG directional emission, we also quantified fluctuations of vesicle orientation. Thus, by combining endocytosis of nanocrystals having a nonlinear response, fast two-photon microscopy, and high-throughput analysis, we are able to finely monitor fast axonal transport in vivo in the brain of a vertebrate and reveal subtle axonal transport alterations. The high spatiotemporal resolution achieved in our model may be relevant to precisely investigate axonal transport impairment associated with disease models.


Asunto(s)
Dineínas , Cinesinas , Animales , Cinesinas/metabolismo , Dineínas/metabolismo , Pez Cebra/metabolismo , Transporte Axonal/genética , Microscopía , Larva/metabolismo , Axones , Microtúbulos/metabolismo , Encéfalo/metabolismo
13.
J Biol Chem ; 285(14): 10252-64, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20154089

RESUMEN

The abnormally folded form of the prion protein (PrP(Sc)) accumulating in nervous and lymphoid tissues of prion-infected individuals can be naturally cleaved to generate a N-terminal-truncated fragment called C2. Information about the identity of the cellular proteases involved in this process and its possible role in prion biology has remained limited and controversial. We investigated PrP(Sc) N-terminal trimming in different cell lines and primary cultured nerve cells, and in the brain and spleen tissue from transgenic mice infected by ovine and mouse prions. We found the following: (i) the full-length to C2 ratio varies considerably depending on the infected cell or tissue. Thus, in primary neurons and brain tissue, PrP(Sc) accumulated predominantly as untrimmed species, whereas efficient trimming occurred in Rov and MovS cells, and in spleen tissue. (ii) Although C2 is generally considered to be the counterpart of the PrP(Sc) proteinase K-resistant core, the N termini of the fragments cleaved in vivo and in vitro can actually differ, as evidenced by a different reactivity toward the Pc248 anti-octarepeat antibody. (iii) In lysosome-impaired cells, the ratio of full-length versus C2 species dramatically increased, yet efficient prion propagation could occur. Moreover, cathepsin but not calpain inhibitors markedly inhibited C2 formation, and in vitro cleavage by cathepsins B and L produced PrP(Sc) fragments lacking the Pc248 epitope, strongly arguing for the primary involvement of acidic hydrolases of the endolysosomal compartment. These findings have implications on the molecular analysis of PrP(Sc) and cell pathogenesis of prion infection.


Asunto(s)
Encéfalo/metabolismo , Proteínas PrPSc/metabolismo , Enfermedades por Prión/metabolismo , Enfermedades por Prión/transmisión , Bazo/metabolismo , Animales , Encéfalo/patología , Calpaína/antagonistas & inhibidores , Calpaína/metabolismo , Catepsina B/antagonistas & inhibidores , Catepsina B/metabolismo , Catepsina L/antagonistas & inhibidores , Catepsina L/metabolismo , Células Cultivadas , Endopeptidasa K/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovinos , Bazo/patología
14.
Neurobiol Dis ; 41(1): 219-25, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20875860

RESUMEN

Prion strains yield specific neuropathological features including spongiform degeneration and deposition patterns of pathological prion protein. Their invariant regional distribution, following variations in the infection route, has led to the proposal that prions replicate preferentially in defined neuro-anatomical areas. The molecular mechanisms underlying this apparent strain-specific neuronal tropism are currently unknown. However, a possible explanation may be that prion replication is relatively innocuous, resulting in long-term propagation, thus masking initial regional distribution variations linked to different infection routes. This "low neurotoxicity" may be imputable either to the rodent model used or the prion strain(s) inoculated. To investigate this possibility, we studied prion pathogenesis in a prototypal short-incubation disease model consisting of 127S scrapie strain propagated in tg338 transgenic mice expressing the VRQ allele of ovine PrP. This prion strain derives from a natural sheep scrapie isolate that was serially transmitted to tg338 mice without any obvious transmission barrier and biologically cloned by limiting dilution. We compared the pathology induced by the peripheral or intracerebral inoculation of 127S strain. Surprisingly, we found that the disease greatly differed in clinical signs, abnormal prion protein levels, and neuropathology among the routes of infection. Secondary transmission performed with brain material from mice inoculated either intracranially or intraperitoneally produced similar neuropathological features. These results therefore indicate that the route of infection can strongly influence the apparent phenotype of a scrapie strain.


Asunto(s)
Transmisión de Enfermedad Infecciosa , Proteínas PrPSc/patogenicidad , Scrapie/genética , Scrapie/patología , Animales , Modelos Animales de Enfermedad , Inyecciones/métodos , Inyecciones Intraperitoneales/métodos , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Neuronas/patología , Fenotipo , Proteínas PrPSc/genética , Proteínas PrPSc/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Scrapie/etiología , Oveja Doméstica
15.
Biochem J ; 431(2): 189-98, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20670217

RESUMEN

TSEs (transmissible spongiform encephalopathies) are neurodegenerative diseases caused by pathogenic isoforms (PrPSc) of the host-encoded PrPc (cellular prion protein). After consumption of contaminated food, PrPSc deposits rapidly accumulate in lymphoid tissues before invasion of the CNS (central nervous system). However, the mechanisms of prion spreading from the periphery to the nervous system are still unclear. In the present study, we investigated the role of DCs (dendritic cells) in the spreading of prion infection to neuronal cells. First, we determined that BMDCs (bone-marrow-derived DCs) rapidly uptake PrPSc after exposure to infected brain homogenate. Next, we observed a progressive catabolism of the internalized prion aggregates. Similar experiments performed with BMDCs isolated from KO (knockout) mice or mice overexpressing PrP (tga20) indicate that both PrPSc uptake and catabolism are independent of PrPc expression in these cells. Finally, using co-cultures of prion-loaded BMDCs and cerebellar neurons, we characterized the transfer of the prion protein and the resulting infection of the neuronal cultures. Interestingly, the transfer of PrPSc was triggered by direct cell-cell contact. As a consequence, BMDCs retained the prion protein when cultured alone, and no transfer to the recipient neurons was observed when a filter separated the two cultures or when neurons were exposed to the BMDC-conditioned medium. Additionally, fixed BMDCs also failed to transfer prion infectivity to neurons, suggesting an active transport of prion aggregates, in accordance with a role of TNTs (tunnelling nanotubes) observed in the co-cultures.


Asunto(s)
Células Dendríticas/metabolismo , Neuronas/metabolismo , Priones/metabolismo , Animales , Células de la Médula Ósea/citología , Células Cultivadas , Células Dendríticas/citología , Endocitosis , Ratones , Ratones Endogámicos C57BL , Neuronas/patología , Proteínas PrPSc/metabolismo , Priones/patogenicidad , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Factores de Tiempo
16.
Front Immunol ; 12: 637399, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33708225

RESUMEN

Innate immune pathways are the first line of cellular defense against pathogen infections ranging from bacteria to Metazoa. These pathways are activated following the recognition of pathogen associated molecular patterns (PAMPs) by membrane and cytosolic pattern recognition receptors. In addition, some of these cellular sensors can also recognize endogenous danger-associated molecular patterns (DAMPs) arising from damaged or dying cells and triggering innate immune responses. Among the cytosolic nucleic acid sensors, the cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS) plays an essential role in the activation of the type I interferon (IFNs) response and the production of pro-inflammatory cytokines. Indeed, upon nucleic acid binding, cGAS synthesizes cGAMP, a second messenger mediating the activation of the STING signaling pathway. The functional conservation of the cGAS-STING pathway during evolution highlights its importance in host cellular surveillance against pathogen infections. Apart from their functions in immunity, cGAS and STING also play major roles in nuclear functions and tumor development. Therefore, cGAS-STING is now considered as an attractive target to identify novel biomarkers and design therapeutics for auto-inflammatory and autoimmune disorders as well as infectious diseases and cancer. Here, we review the current knowledge about the structure of cGAS and the evolution from bacteria to Metazoa and present its main functions in defense against pathogens and cancer, in connection with STING. The advantages and limitations of in vivo models relevant for studying the cGAS-STING pathway will be discussed for the notion of species specificity and in the context of their integration into therapeutic screening assays targeting cGAG and/or STING.


Asunto(s)
ADN/genética , Enfermedades del Sistema Inmune/metabolismo , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/metabolismo , Animales , ADN/inmunología , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Espacio Intracelular , Proteínas de la Membrana/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal
17.
Viruses ; 13(2)2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33525646

RESUMEN

BACKGROUND: Respiratory Syncytial Virus (RSV) is the major cause of severe acute respiratory tract illness in young children worldwide and a main pathogen for the elderly and immune-compromised people. In the absence of vaccines or effective treatments, a better characterization of the pathogenesis of RSV infection is required. To date, the pathophysiology of the disease and its diagnosis has mostly relied on chest X-ray and genome detection in nasopharyngeal swabs. The development of new imaging approaches is instrumental to further the description of RSV spread, virus-host interactions and related acute respiratory disease, at the level of the entire lung. METHODS: By combining tissue clearing, 3D microscopy and image processing, we developed a novel visualization tool of RSV infection in undissected mouse lungs. RESULTS: Whole tissue analysis allowed the identification of infected cell subtypes, based on both morphological traits and position within the cellular network. Furthermore, 3D imaging was also valuable to detect the cytoplasmic viral factories, also called inclusion bodies, a hallmark of RSV infection. CONCLUSIONS: Whole lung clearing and 3D deep imaging represents an unprecedented visualization method of infected lungs to allow insight into RSV pathophysiology and improve the 2D histology analyses.


Asunto(s)
Imagenología Tridimensional , Pulmón/patología , Pulmón/virología , Virus Sincitial Respiratorio Humano/fisiología , Animales , Modelos Animales de Enfermedad , Células Epiteliales/virología , Cuerpos de Inclusión Viral/patología , Ratones , Infecciones por Virus Sincitial Respiratorio/patología , Infecciones por Virus Sincitial Respiratorio/virología , Replicación Viral
18.
J Virol ; 83(19): 10256-63, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19625402

RESUMEN

It was recently reported that human immunodeficiency virus type 1 (HIV-1) Vpr induced the proteasomal degradation of the nuclear UNG2 enzyme for efficient virus replication. We confirm here that HIV-1 infection and Vpr expression reduce the level of endogenous UNG2, but this effect is not reverted by treatment with the proteasome inhibitor MG132. Moreover, this reduction is not mediated by Vpr binding to UNG2 and is independent of the Vpr-induced G(2) arrest. Finally, we show that Vpr influences the UNG2 promoter without affecting UNG1 gene expression. These data indicate that the Vpr-induced decrease of UNG2 level is mainly related to a transcriptional effect.


Asunto(s)
ADN Glicosilasas/biosíntesis , Regulación Viral de la Expresión Génica , Transcripción Genética , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo , Ciclo Celular , Núcleo Celular/metabolismo , Fase G2 , VIH-1/genética , Células HeLa , Humanos , Leupeptinas/farmacología , Microscopía Fluorescente/métodos , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Replicación Viral
19.
J Mol Biol ; 432(20): 5529-5543, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32860771

RESUMEN

Unresolved inflammation fosters and supports a wide range of human pathologies. There is growing evidence for a role played by cytosolic nucleic acids in initiating and supporting pathological chronic inflammation. In particular, the cGAS-STING pathway has emerged as central to the mounting of nucleic acid-dependent type I interferon responses, leading to the identification of small-molecule modulators of STING that have raised clinical interest. However, several new challenges have emerged, representing potential obstacles to efficient clinical translation. Indeed, the current literature underscores that nucleic acid-induced inflammatory responses are subjected to several layers of regulation, further suggesting complex coordination at the cell-type, tissue or organism level. Untangling the underlying processes is paramount to the identification of specific therapeutic strategies targeting deleterious inflammation. Herein, we present an overview of human pathologies presenting with deregulated interferon levels and with accumulation of cytosolic nucleic acids. We focus on the central role of the STING adaptor protein in these pathologies and discuss how in vivo models have forged our current understanding of nucleic acid immunity. We present our opinion on the advantages and limitations of zebrafish and mice models to highlight their complementarity for the study of inflammatory human pathologies and the development of therapeutics. Finally, we discuss high-throughput screening strategies that generate multi-parametric datasets that allow integrative analysis of heterogeneous information (imaging and omics approaches). These approaches are likely to structure the future of screening strategies for the treatment of human pathologies.


Asunto(s)
Inmunidad , Modelos Animales , Ácidos Nucleicos/inmunología , Animales , Antivirales/farmacología , Citosol/metabolismo , ADN Viral/análisis , Evaluación Preclínica de Medicamentos , Evolución Molecular , Ensayos Analíticos de Alto Rendimiento/métodos , Inmunidad Innata , Inflamación/inmunología , Inflamación/metabolismo , Interferón Tipo I/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Mutación , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Transducción de Señal
20.
Viruses ; 11(3)2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30917538

RESUMEN

The overarching structure of the type I interferon (IFN) system is conserved across vertebrates. However, the variable numbers of whole genome duplication events during fish evolution offer opportunities for the expansion, diversification, and new functionalization of the genes that are involved in antiviral immunity. In this review, we examine how fish models provide new insights about the implication of virus-driven inflammation in immunity and hematopoiesis. Mechanisms that have been discovered in fish, such as the strong adjuvant effect of type I IFN that is used with DNA vaccination, constitute good models to understand how virus-induced inflammatory mechanisms can interfere with adaptive responses. We also comment on new discoveries regarding the role of pathogen-induced inflammation in the development and guidance of hematopoietic stem cells in zebrafish. These findings raise issues about the potential interferences of viral infections with the establishment of the immune system. Finally, the recent development of genome editing provides new opportunities to dissect the roles of the key players involved in the antiviral response in fish, hence enhancing the power of comparative approaches.


Asunto(s)
Modelos Animales de Enfermedad , Interferón Tipo I/inmunología , Transducción de Señal/inmunología , Virosis/veterinaria , Pez Cebra/inmunología , Pez Cebra/virología , Adyuvantes Inmunológicos , Animales , Edición Génica , Inmunidad Innata , Inflamación , Virosis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA