Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 149
Filtrar
1.
Antonie Van Leeuwenhoek ; 117(1): 39, 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38388985

RESUMEN

Melioidosis, a human infectious disease with a high mortality rate in many tropical countries, is caused by the pathogen Burkholderia pseudomallei (B. pseudomallei). The function of the B. pseudomallei sigma S (RpoS) transcription factor in survival during the stationary growth phase and conditions of oxidative stress is well documented. Besides the rpoS, bioinformatics analysis of B. pseudomallei genome showed the existence of two rpoN genes, named rpoN1 and rpoN2. In this study, by using the mouse macrophage cell line RAW264.7 as a model of infection, the involvement of B. pseudomallei RpoS and RpoN2 in the invasion, intracellular survival leading to the reduction in multinucleated giant cell (MNGC) formation of RAW264.7 cell line were illustrated. We have demonstrated that the MNGC formation of RAW264.7 cell was dependent on a certain number of intracellular bacteria (at least 5 × 104). In addition, the same MNGC formation (15%) observed in RAW264.7 cells infected with either B. pseudomallei wild type with multiplicity of infection (MOI) 2 or RpoN2 mutant (∆rpoN2) with MOI 10 or RpoS mutant (∆rpoS) with MOI 100. The role of B. pseudomallei RpoS and RpoN2 in the regulation of type III secretion system on bipB-bipC gene expression was also illustrated in this study.


Asunto(s)
Burkholderia pseudomallei , Melioidosis , Animales , Ratones , Humanos , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/metabolismo , Línea Celular , Melioidosis/microbiología , Macrófagos/metabolismo , Células Gigantes/metabolismo , Células Gigantes/microbiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo
2.
Science ; 381(6662): eabq5202, 2023 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-37676943

RESUMEN

Kupffer cells (KCs) are localized in liver sinusoids but extend pseudopods to parenchymal cells to maintain their identity and serve as the body's central bacterial filter. Liver cirrhosis drastically alters vascular architecture, but how KCs adapt is unclear. We used a mouse model of liver fibrosis and human tissue to examine immune adaptation. Fibrosis forced KCs to lose contact with parenchymal cells, down-regulating "KC identity," which rendered them incapable of clearing bacteria. Commensals stimulated the recruitment of monocytes through CD44 to a spatially distinct vascular compartment. There, recruited monocytes formed large aggregates of multinucleated cells (syncytia) that expressed phenotypical KC markers and displayed enhanced bacterial capture ability. Syncytia formed via CD36 and were observed in human cirrhosis as a possible antimicrobial defense that evolved with fibrosis.


Asunto(s)
Infecciones de Transmisión Sanguínea , Células Gigantes , Macrófagos del Hígado , Cirrosis Hepática , Animales , Humanos , Ratones , Células Gigantes/inmunología , Células Gigantes/microbiología , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/microbiología , Cirrosis Hepática/inmunología , Cirrosis Hepática/microbiología , Cirrosis Hepática/patología , Infecciones de Transmisión Sanguínea/inmunología , Modelos Animales de Enfermedad
3.
mBio ; 12(4): e0205421, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34399626

RESUMEN

Inflammasomes are cytosolic multiprotein signaling complexes that are activated upon pattern recognition receptor-mediated recognition of pathogen-derived ligands or endogenous danger signals. Their assembly activates the downstream inflammatory caspase-1 and caspase-4/5 (human) or caspase-11 (mouse), which induces cytokine release and pyroptotic cell death through the cleavage of the pore-forming effector gasdermin D. Pathogen detection by host cells also results in the production and release of interferons (IFNs), which fine-tune inflammasome-mediated responses. IFN-induced guanylate-binding proteins (GBPs) have been shown to control the activation of the noncanonical inflammasome by recruiting caspase-4 on the surface of cytosolic Gram-negative bacteria and promoting its interaction with lipopolysaccharide (LPS). The Gram-negative opportunistic bacterial pathogen Burkholderia thailandensis infects epithelial cells and macrophages and hijacks the host actin polymerization machinery to spread into neighboring cells. This process causes host cell fusion and the formation of so-called multinucleated giant cells (MNGCs). Caspase-1- and IFN-regulated caspase-11-mediated inflammasome pathways play an important protective role against B. thailandensis in mice, but little is known about the role of IFNs and inflammasomes during B. thailandensis infection of human cells, particularly epithelial cells. Here, we report that IFN-γ priming of human epithelial cells restricts B. thailandensis-induced MNGC formation in a GBP1-dependent manner. Mechanistically, GBP1 does not promote bacteriolysis or impair actin-based bacterial motility but acts by inducing caspase-4-dependent pyroptosis of the infected cell. In addition, we show that IFN-γ priming of human primary macrophages confers a more efficient antimicrobial effect through inflammasome activation, further confirming the important role that interferon signaling plays in restricting Burkholderia replication and spread. IMPORTANCE The Gram-negative bacteria of the Burkholderia species are associated with human diseases ranging from pneumonia to life-threatening melioidosis. Upon infection through inhalation, ingestion, or the percutaneous route, these bacteria can spread and establish granuloma-like lesions resulting from the fusion of host cells to form multinucleated giant cells (MNGCs). Burkholderia resistance to several antibiotics highlights the importance to better understand how the innate immune system controls infections. Here, we report that interferons protect human epithelial cells against Burkholderia-induced MNGC formation, specifically through the action of the interferon-induced GBP1 protein. Mechanistically, GBP1 acts by inducing caspase-4-dependent cell death through pyroptosis, allowing the infected cells to be quickly eliminated before bacterial spread and the formation of MNGCs. This study provides evidence that interferon-induced innate immune activation, through GBP1 and caspase-4, confers protection against Burkholderia infection, potentially opening new perspectives for therapeutic approaches.


Asunto(s)
Burkholderia/inmunología , Células Epiteliales/microbiología , Proteínas de Unión al GTP/genética , Células Gigantes/microbiología , Inflamasomas/inmunología , Interferón gamma/metabolismo , Burkholderia/química , Burkholderia/genética , Fusión Celular , Citosol , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Proteínas de Unión al GTP/metabolismo , Células Gigantes/fisiología , Células HeLa , Humanos , Inflamasomas/genética , Interferón gamma/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/fisiología , Fagocitosis , Transducción de Señal/inmunología
4.
Nat Commun ; 12(1): 2027, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33795674

RESUMEN

The immune response to mycobacteria is characterized by granuloma formation, which features multinucleated giant cells as a unique macrophage type. We previously found that multinucleated giant cells result from Toll-like receptor-induced DNA damage and cell autonomous cell cycle modifications. However, the giant cell progenitor identity remained unclear. Here, we show that the giant cell-forming potential is a particular trait of monocyte progenitors. Common monocyte progenitors potently produce cytokines in response to mycobacteria and their immune-active molecules. In addition, common monocyte progenitors accumulate cholesterol and lipids, which are prerequisites for giant cell transformation. Inducible monocyte progenitors are so far undescribed circulating common monocyte progenitor descendants with high giant cell-forming potential. Monocyte progenitors are induced in mycobacterial infections and localize to granulomas. Accordingly, they exhibit important immunological functions in mycobacterial infections. Moreover, their signature trait of high cholesterol metabolism may be piggy-backed by mycobacteria to create a permissive niche.


Asunto(s)
Citocinas/inmunología , Células Gigantes/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Células Madre/inmunología , Animales , Células Cultivadas , Citocinas/metabolismo , Femenino , Células Gigantes/metabolismo , Células Gigantes/microbiología , Granuloma/inmunología , Granuloma/metabolismo , Humanos , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Monocitos/metabolismo , Monocitos/microbiología , Mycobacterium/inmunología , Mycobacterium/fisiología , Células Madre/metabolismo , Células Madre/microbiología
5.
mBio ; 12(2)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33688010

RESUMEN

Cryptococcus neoformans is a ubiquitous, opportunistic fungal pathogen that kills almost 200,000 people worldwide each year. It is acquired when mammalian hosts inhale the infectious propagules; these are deposited in the lung and, in the context of immunocompromise, may disseminate to the brain and cause lethal meningoencephalitis. Once inside the host, C. neoformans undergoes a variety of adaptive processes, including secretion of virulence factors, expansion of a polysaccharide capsule that impedes phagocytosis, and the production of giant (Titan) cells. The transcription factor Pdr802 is one regulator of these responses to the host environment. Expression of the corresponding gene is highly induced under host-like conditions in vitro and is critical for C. neoformans dissemination and virulence in a mouse model of infection. Direct targets of Pdr802 include the quorum sensing proteins Pqp1, Opt1, and Liv3; the transcription factors Stb4, Zfc3, and Bzp4, which regulate cryptococcal brain infectivity and capsule thickness; the calcineurin targets Had1 and Crz1, important for cell wall remodeling and C. neoformans virulence; and additional genes related to resistance to host temperature and oxidative stress, and to urease activity. Notably, cryptococci engineered to lack Pdr802 showed a dramatic increase in Titan cells, which are not phagocytosed and have diminished ability to directly cross biological barriers. This explains the limited dissemination of pdr802 mutant cells to the central nervous system and the consequently reduced virulence of this strain. The role of Pdr802 as a negative regulator of Titan cell formation is thus critical for cryptococcal pathogenicity.IMPORTANCE The pathogenic yeast Cryptococcus neoformans presents a worldwide threat to human health, especially in the context of immunocompromise, and current antifungal therapy is hindered by cost, limited availability, and inadequate efficacy. After the infectious particle is inhaled, C. neoformans initiates a complex transcriptional program that integrates cellular responses and enables adaptation to the host lung environment. Here, we describe the role of the transcription factor Pdr802 in the response to host conditions and its impact on C. neoformans virulence. We identified direct targets of Pdr802 and also discovered that it regulates cellular features that influence movement of this pathogen from the lung to the brain, where it causes fatal disease. These findings significantly advance our understanding of a serious disease.


Asunto(s)
Cryptococcus neoformans/genética , Cryptococcus neoformans/patogenicidad , Proteínas Fúngicas/genética , Regulación Fúngica de la Expresión Génica/genética , Células Gigantes/fisiología , Interacciones Huésped-Patógeno , Factores de Transcripción/genética , Animales , Femenino , Proteínas Fúngicas/metabolismo , Eliminación de Gen , Células Gigantes/microbiología , Ratones , Ratones Endogámicos BALB C , Factores de Transcripción/metabolismo , Factores de Virulencia/metabolismo
6.
Sci Rep ; 10(1): 17972, 2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33087788

RESUMEN

Tetraspanins are four-span transmembrane proteins of host cells that facilitate infections by many pathogens. Burkholderia pseudomallei is an intracellular bacterium and the causative agent of melioidosis, a severe disease in tropical regions. This study investigated the role of tetraspanins in B. pseudomallei infection. We used flow cytometry to determine tetraspanins CD9, CD63, and CD81 expression on A549 and J774A.1 cells. Their roles in B. pseudomallei infection were investigated in vitro using monoclonal antibodies (MAbs) and recombinant large extracellular loop (EC2) proteins to pretreat cells before infection. Knockout of CD9 and CD81 in cells was performed using CRISPR Cas9 to confirm the role of tetraspanins. Pretreatment of A549 cells with MAb against CD9 and CD9-EC2 significantly enhanced B. pseudomallei internalization, but MAb against CD81 and CD81-EC2 inhibited MNGC formation. Reduction of MNGC formation was consistently observed in J774.A1 cells pretreated with MAbs specific to CD9 and CD81 and with CD9-EC2 and CD81-EC2. Data from knockout experiments confirmed that CD9 enhanced bacterial internalization and that CD81 inhibited MNGC formation. Our data indicate that tetraspanins are host cellular factors that mediated internalization and membrane fusion during B. pseudomallei infection. Tetraspanins may be the potential therapeutic targets for melioidosis.


Asunto(s)
Burkholderia pseudomallei/patogenicidad , Fusión Celular , Melioidosis/microbiología , Fagocitos/fisiología , Tetraspaninas/fisiología , Células A549 , Anticuerpos Monoclonales , Sistemas CRISPR-Cas , Células Cultivadas , Células Gigantes/microbiología , Humanos , Melioidosis/terapia , Tetraspanina 28 , Tetraspanina 29 , Tetraspaninas/metabolismo
7.
Infect Immun ; 89(1)2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33106293

RESUMEN

Burkholderia pseudomallei is a Gram-negative bacterium and the causative agent of melioidosis. Despite advances in our understanding of the disease, B. pseudomallei poses a significant health risk, especially in regions of endemicity, where treatment requires prolonged antibiotic therapy. Even though the respiratory and percutaneous routes are well documented and considered the main ways to acquire the pathogen, the gastrointestinal tract is believed to be an underreported and underrecognized route of infection. In the present study, we describe the development of in vitro and in vivo models to study B. pseudomallei gastrointestinal infection. Further, we report that the type 6 secretion system (T6SS) and type 1 fimbriae are important virulence factors required for gastrointestinal infection. Using a human intestinal epithelial cell line and mouse primary intestinal epithelial cells (IECs), we demonstrated that B. pseudomallei adheres, invades, and forms multinucleated giant cells, ultimately leading to cell toxicity. We demonstrated that mannose-sensitive type 1 fimbria is involved in the initial adherence of B. pseudomallei to IECs, although the impact on full virulence was limited. Finally, we also showed that B. pseudomallei requires a functional T6SS for full virulence, bacterial dissemination, and lethality in mice infected by the intragastric route. Overall, we showed that B. pseudomallei is an enteric pathogen and that type 1 fimbria is important for B. pseudomallei intestinal adherence, and we identify a new role for T6SS as a key virulence factor in gastrointestinal infection. These studies highlight the importance of gastrointestinal melioidosis as an understudied route of infection and open a new avenue for the pathogenesis of B. pseudomallei.


Asunto(s)
Burkholderia pseudomallei/fisiología , Gastroenteritis/microbiología , Melioidosis/microbiología , Factores de Virulencia/genética , Animales , Adhesión Bacteriana/genética , Burkholderia pseudomallei/patogenicidad , Modelos Animales de Enfermedad , Fimbrias Bacterianas/fisiología , Regulación Bacteriana de la Expresión Génica , Células Gigantes/microbiología , Células Gigantes/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Ratones , Sistemas de Secreción Tipo VI , Virulencia/genética
8.
PLoS Negl Trop Dis ; 14(9): e0008590, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32991584

RESUMEN

BACKGROUND: Burkholderia pseudomallei is an environmental bacterium that causes melioidosis. A facultative intracellular pathogen, B. pseudomallei can induce multinucleated giant cells (MNGCs) leading to plaque formation in vitro. B. pseudomallei can switch colony morphotypes under stress conditions. In addition, different isolates have been reported to have varying virulence in vivo, but genomic evolution and the relationship with plaque formation is poorly understood. METHODOLOGY/PRINCIPLE FINDINGS: To gain insights into genetic underpinnings of virulence of B. pseudomallei, we screened plaque formation of 52 clinical isolates and 11 environmental isolates as well as 4 isogenic morphotype isolates of B. pseudomallei strains K96243 (types II and III) and 153 (types II and III) from Thailand in A549 and HeLa cells. All isolates except one environmental strain (A4) and K96243 morphotype II were able to induce plaque formation in both cell lines. Intracellular growth assay and confocal microscopy analyses demonstrated that the two plaque-forming-defective isolates were also impaired in intracellular replication, actin polymerization and MNGC formation in infected cells. Whole genome sequencing analysis and PCR revealed that both isolates had a large genomic loss on the same region in chromosome 2, which included Bim cluster, T3SS-3 and T6SS-5 genes. CONCLUSIONS/SIGNIFICANCE: Our plaque screening and genomic studies revealed evidence of impairment in plaque formation in environmental isolates of B. pseudomallei that is associated with large genomic loss of genes important for intracellular multiplication and MNGC formation. These findings suggest that the genomic and phenotypic differences of environmental isolates may be associated with clinical infection.


Asunto(s)
Burkholderia pseudomallei/genética , Burkholderia pseudomallei/aislamiento & purificación , Genoma Bacteriano/genética , Células Gigantes/microbiología , Macrófagos/microbiología , Células A549 , Adulto , Anciano , Burkholderia pseudomallei/patogenicidad , Línea Celular Tumoral , Femenino , Eliminación de Gen , Células HeLa , Humanos , Masculino , Melioidosis/microbiología , Melioidosis/patología , Técnicas Microbiológicas , Persona de Mediana Edad , Estudios Prospectivos , Secuenciación Completa del Genoma , Adulto Joven
9.
Med Microbiol Immunol ; 209(4): 473-487, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32253503

RESUMEN

Burkholderia pseudomallei is the causative agent of melioidosis, a disease with high morbidity that is endemic in South East Asia and northern Australia. An unusual feature of the bacterium is its ability to induce multinucleated giant cell formation (MNGC), which appears to be related to bacterial pathogenicity. The mechanism of MNGC formation is not fully understood, but host cell factors as well as known bacterial virulence determinants are likely to contribute. Since members of the tetraspanin family of membrane proteins are involved in various types of cell:cell fusion, their role in MNGC formation induced by Burkholderia thailandensis, a mildly pathogenic species closely related to B. pseudomallei, was investigated. The effect of antibodies to tetraspanins CD9, CD81, and CD63 in MNGC formation induced by B. thailandensis in infected mouse J774.2 and RAW macrophage cell lines was assessed along with that of recombinant proteins corresponding to the large extracellular domain (EC2) of the tetraspanins. B. thailandensis-induced fusion was also examined in macrophages derived from CD9 null and corresponding WT mice, and in J774.2 macrophages over-expressing CD9. Antibodies to CD9 and CD81 promoted MNGC formation induced by B. thailandensis, whereas EC2 proteins of CD9, CD81, and CD63 inhibited MNGC formation. Enhanced MNGC formation was observed in CD9 null macrophages, whereas a decrease in MNGC formation was associated with overexpression of CD9. Overall our findings show that tetraspanins are involved in MNGC formation induced by B. thailandensis and by implication, B. pseudomallei, with CD9 and CD81 acting as negative regulators of this process.


Asunto(s)
Burkholderia , Fusión Celular , Células Gigantes/metabolismo , Macrófagos/microbiología , Tetraspaninas/metabolismo , Animales , Burkholderia pseudomallei , Línea Celular , Células Gigantes/microbiología , Melioidosis/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Recombinantes/metabolismo , Tetraspanina 28/metabolismo , Tetraspanina 29/metabolismo , Tetraspanina 30/metabolismo
10.
PLoS Pathog ; 16(3): e1008364, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32150572

RESUMEN

Innate immunity responds to pathogens by producing alarm signals and activating pathways that make host cells inhospitable for pathogen replication. The intracellular bacterium Burkholderia thailandensis invades the cytosol, hijacks host actin, and induces cell fusion to spread to adjacent cells, forming multinucleated giant cells (MNGCs) which promote bacterial replication. We show that type I interferon (IFN) restricts macrophage MNGC formation during B. thailandensis infection. Guanylate-binding proteins (GBPs) expressed downstream of type I IFN were required to restrict MNGC formation through inhibition of bacterial Arp2/3-dependent actin motility during infection. GTPase activity and the CAAX prenylation domain were required for GBP2 recruitment to B. thailandensis, which restricted bacterial actin polymerization required for MNGC formation. Consistent with the effects in in vitro macrophages, Gbp2-/-, Gbp5-/-, GbpChr3-KO mice were more susceptible to intranasal infection with B. thailandensis than wildtype mice. Our findings reveal that IFN and GBPs play a critical role in restricting cell-cell fusion and bacteria-induced pathology during infection.


Asunto(s)
Infecciones por Burkholderia/inmunología , Burkholderia/inmunología , Proteínas de Unión al GTP/inmunología , Células Gigantes/inmunología , Macrófagos/inmunología , Enfermedades Nasales/inmunología , Prenilación de Proteína/inmunología , Animales , Infecciones por Burkholderia/genética , Infecciones por Burkholderia/patología , Fusión Celular , Proteínas de Unión al GTP/genética , Células Gigantes/microbiología , Células Gigantes/patología , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Noqueados , Enfermedades Nasales/genética , Enfermedades Nasales/microbiología , Enfermedades Nasales/patología
11.
Nat Commun ; 10(1): 3831, 2019 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-31444339

RESUMEN

When injured, fish release an alarm substance (Schreckstoff) that elicits fear in members of their shoal. Although Schreckstoff has been proposed to be produced by club cells in the skin, several observations indicate that these giant cells function primarily in immunity. Previous data indicate that the alarm substance can be isolated from mucus. Here we show that mucus, as well as bacteria, are transported from the external surface into club cells, by cytoplasmic transfer or invasion of cells, including neutrophils. The presence of bacteria inside club cells raises the possibility that the alarm substance may contain a bacterial component. Indeed, lysate from a zebrafish Staphylococcus isolate is sufficient to elicit alarm behaviour, acting in concert with a substance from fish. These results suggest that Schreckstoff, which allows one individual to unwittingly change the emotional state of the surrounding population, derives from two kingdoms and is associated with processes that protect the host from bacteria.


Asunto(s)
Comunicación Animal , Piel/metabolismo , Staphylococcus/metabolismo , Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Miedo/fisiología , Células Gigantes/metabolismo , Células Gigantes/microbiología , Microscopía Intravital , Moco/citología , Moco/metabolismo , Moco/microbiología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Imagen Óptica , Reflejo de Sobresalto/fisiología , Piel/citología , Piel/microbiología , Simbiosis/fisiología , Pez Cebra/lesiones , Pez Cebra/microbiología
12.
Neurochirurgie ; 64(6): 431-433, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30413280

RESUMEN

INTRODUCTION: Aneurysmal bone cysts (ABCs) are rare, rapidly expansile, benign, vascular lesions capable of causing local bone destruction. The majority of cases present as multi-cystic lytic lesions (with solid-variant ABCs representing<10% of all presentations) of the long bones or vertebrae, rarely occurring in the head/neck region. CLINICAL CASE: A 44-year-old female presented with nine days of worsening pain, ptosis and proptosis in the right eye. CT and MR imaging revealed a 3.2cm extra-axial multiloculated right frontal lobe mass in the orbit with fluid-fluid levels secondary to layering of solid blood components. A right craniotomy was performed and the lesion was resected piecemeal with subsequent high speed burring to remove residual tissue. Histological evaluation revealed spindle and giant cell infiltration of the bone without vascular channels. Based on these findings, the lesion was diagnosed as a solid-variant orbital ABC without paranasal sinus involvement. The patient recovered fully with no residual symptoms. CONCLUSION: This case report details a rare presentation of ABC (solid-variant presenting outside of the vertebrae/long bones) with discussion concerning possible treatment modalities and guidance for follow-up.


Asunto(s)
Quistes Óseos Aneurismáticos/patología , Quistes Óseos Aneurismáticos/cirugía , Craneotomía , Órbita/cirugía , Columna Vertebral/cirugía , Adulto , Quistes Óseos Aneurismáticos/diagnóstico , Craneotomía/métodos , Femenino , Células Gigantes/microbiología , Humanos , Imagen por Resonancia Magnética/métodos , Cuello/patología , Cuello/cirugía , Dolor/fisiopatología , Dolor/cirugía
13.
Mol Plant Pathol ; 19(7): 1690-1704, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29240311

RESUMEN

Photosynthetic efficiency and redox homeostasis are important for plant physiological processes during regular development as well as defence responses. The second-stage juveniles of Heterodera schachtii induce syncytial feeding sites in host roots. To ascertain whether the development of syncytia alters photosynthesis and the metabolism of reactive oxygen species (ROS), chlorophyll a fluorescence measurements and antioxidant responses were studied in Arabidopsis thaliana shoots on the day of inoculation and at 3, 7 and 15 days post-inoculation (dpi). Nematode parasitism caused an accumulation of superoxide and hydrogen peroxide molecules in the shoots of infected plants at 3 dpi, probably as a result of the observed down-regulation of antioxidant enzymes. These changes were accompanied by an increase in RNA and lipid oxidation markers. The activities of antioxidant enzymes were found to be enhanced on infection at 7 and 15 dpi, and the content of anthocyanins was elevated from 3 dpi. The fluorescence parameter Rfd , defining plant vitality and the photosynthetic capacity of leaves, decreased by 11% only at 7 dpi, and non-photochemical quenching (NPQ), indicating the effectiveness of photoprotection mechanisms, was about 16% lower at 3 and 7 dpi. As a result of infection, the ultrastructure of chloroplasts was changed (large starch grains and plastoglobules), and more numerous and larger peroxisomes were observed in the mesophyll cells of leaves. We postulate that the joint action of antioxidant enzymes/molecules and photochemical mechanisms leading to the maintenance of photosynthetic efficiency promotes the fine-tuning of the infected plants to oxidative stress induced by parasitic cyst nematodes.


Asunto(s)
Fotosíntesis/fisiología , Enfermedades de las Plantas/parasitología , Especies Reactivas de Oxígeno/metabolismo , Tylenchoidea/patogenicidad , Animales , Arabidopsis/metabolismo , Arabidopsis/microbiología , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Beta vulgaris/metabolismo , Beta vulgaris/microbiología , Regulación de la Expresión Génica de las Plantas , Células Gigantes/metabolismo , Células Gigantes/microbiología
14.
EMBO Rep ; 18(12): 2144-2159, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29097394

RESUMEN

Immunity to mycobacteria involves the formation of granulomas, characterized by a unique macrophage (MΦ) species, so-called multinucleated giant cells (MGC). It remains unresolved whether MGC are beneficial to the host, that is, by prevention of bacterial spread, or whether they promote mycobacterial persistence. Here, we show that the prototypical antimycobacterial molecule nitric oxide (NO), which is produced by MGC in excessive amounts, is a double-edged sword. Next to its antibacterial capacity, NO propagates the transformation of MΦ into MGC, which are relatively permissive for mycobacterial persistence. The mechanism underlying MGC formation involves NO-induced DNA damage and impairment of p53 function. Moreover, MGC have an unsurpassed potential to engulf mycobacteria-infected apoptotic cells, which adds a further burden to their antimycobacterial capacity. Accordingly, mycobacteria take paradoxical advantage of antimicrobial cellular efforts by driving effector MΦ into a permissive MGC state.


Asunto(s)
Células Gigantes/microbiología , Macrófagos/fisiología , Mycobacterium/metabolismo , Óxido Nítrico/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Daño del ADN , Genes p53/fisiología , Células Gigantes/metabolismo , Humanos , Macrófagos/microbiología , Ratones , Mycobacterium/inmunología , Óxido Nítrico/biosíntesis
15.
Infect Immun ; 85(10)2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28760929

RESUMEN

The human pathogen Burkholderia pseudomallei and the related species Burkholderia thailandensis are facultative intracellular bacteria characterized by the ability to escape into the cytosol of the host cell and to stimulate the formation of multinucleated giant cells (MNGCs). MNGC formation is induced via an unknown mechanism by bacterial type VI secretion system 5 (T6SS-5), which is an essential virulence factor in both species. Despite the vital role of the intracellular life cycle in the pathogenesis of the bacteria, the range of host cell types permissive for initiation and completion of the intracellular cycle is poorly defined. In the present study, we used several different types of human primary cells to evaluate bacterial entry, intracellular survival, and MNGC formation. We report the capacity of B. pseudomallei to enter, efficiently replicate in, and mediate MNGC formation of vein endothelial and bronchial epithelial cells, indicating that the T6SS-5 is important in the host-pathogen interaction in these cells. Furthermore, we show that B. pseudomallei invades fibroblasts and keratinocytes and survives inside these cells as well as in monocyte-derived macrophages and neutrophils for at least 17 h postinfection; however, MNGC formation is not induced in these cells. In contrast, infection of mixed neutrophils and RAW264.7 macrophages with B. thailandensis stimulated the formation of heterotypic MNGCs in a T6SS-5-dependent manner. In summary, the ability of the bacteria to enter and survive as well as induce MNGC formation in certain host cells may contribute to the pathogenesis observed in B. pseudomallei infection.


Asunto(s)
Burkholderia pseudomallei/fisiología , Células Gigantes/microbiología , Interacciones Huésped-Patógeno , Macrófagos/microbiología , Fagocitos/microbiología , Animales , Bronquios/citología , Bronquios/microbiología , Burkholderia pseudomallei/crecimiento & desarrollo , Burkholderia pseudomallei/patogenicidad , Línea Celular , Células Cultivadas , Citosol/microbiología , Células Endoteliales/microbiología , Células Epiteliales/microbiología , Fibroblastos/microbiología , Humanos , Queratinocitos/microbiología , Ratones , Neutrófilos/microbiología , Sistemas de Secreción Tipo VI/metabolismo , Virulencia
16.
Nat Microbiol ; 1(11): 16144, 2016 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-27782144

RESUMEN

The growth of pathogens is dictated by their interactions with the host environment1. Obligate intracellular pathogens undergo several cellular decisions as they progress through their life cycles inside host cells2. We have studied this process for microsporidian species in the genus Nematocida as they grew and developed inside their co-evolved animal host, Caenorhabditis elegans3-5. We found that microsporidia can restructure multicellular host tissues into a single contiguous multinucleate cell. In particular, we found that all three Nematocida species we studied were able to spread across the cells of C. elegans tissues before forming spores, with two species causing syncytial formation in the intestine and one species causing syncytial formation in the muscle. We also found that the decision to switch from replication to differentiation in Nematocida parisii was altered by the density of infection, suggesting that environmental cues influence the dynamics of the pathogen life cycle. These findings show how microsporidia can maximize the use of host space for growth and that environmental cues in the host can regulate a developmental switch in the pathogen.


Asunto(s)
Caenorhabditis elegans/microbiología , Células Gigantes/microbiología , Interacciones Huésped-Patógeno , Microsporidios/fisiología , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/fisiología , Citoplasma/microbiología , Intestinos/microbiología , Microsporidios/clasificación , Microsporidios/crecimiento & desarrollo , Músculos/microbiología , Filogenia
17.
Curr Opin Microbiol ; 29: 94-103, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26803404

RESUMEN

The Burkholderia genus contains a group of soil-dwelling Gram-negative organisms that are prevalent in warm and humid climates. Two species in particular are able to cause disease in animals, B. mallei primarily infects Equus spp. and B. pseudomallei (BPS), that is able to cause potentially life-threatening disease in humans. BPS is naturally resistant to many antibiotics and there is no vaccine available. Although not a specialised human pathogen, BPS possesses a large genome and many virulence traits that allow it to adapt and survive very successfully in the human host. Key to this survival is the ability of BPS to replicate intracellularly. In this review we highlight recent advances in our understanding of the intracellular survival of BPS, including how it overcomes host immune defenses and other challenges to establish its niche and then spread the infection. Knowledge of these mechanisms increases our capacity for therapeutic interventions against a well-armed foe.


Asunto(s)
Burkholderia pseudomallei/crecimiento & desarrollo , Burkholderia pseudomallei/inmunología , Citoplasma/microbiología , Interacciones Huésped-Patógeno , Melioidosis/microbiología , Actinas/metabolismo , Animales , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/patogenicidad , Replicación del ADN , Células Gigantes/microbiología , Humanos , Melioidosis/terapia , Sistemas de Secreción Tipo VI/metabolismo , Virulencia/genética , Factores de Virulencia/genética , Factores de Virulencia/fisiología
18.
J Cell Biochem ; 117(1): 132-43, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26084267

RESUMEN

Chlamydia trachomatis, the leading cause of bacterial sexually transmitted infections, disrupts cytokinesis and causes significant multinucleation in host cells. Here, we demonstrate that multinuclear cells that result from unsuccessful cell division contain significantly higher Golgi content, an important source of lipids for chlamydiae. Using immunofluorescence and fluorescent live cell imaging, we show that C. trachomatis in multinuclear cells indeed intercept Golgi-derived lipid faster than in mononuclear cells. Moreover, multinuclear cells enhance C. trachomatis inclusion growth and infectious particle formation. Together, these results indicate that C. trachomatis robustly position inclusions to the cell equator to disrupt host cell division in order to acquire host Golgi-derived lipids more quickly in multinucleated progeny cells.


Asunto(s)
Chlamydia trachomatis/patogenicidad , Citocinesis/fisiología , Células Gigantes/microbiología , División Celular/fisiología , Línea Celular , Aparato de Golgi/metabolismo , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Metafase/fisiología
20.
PLoS One ; 9(6): e100763, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24955832

RESUMEN

Chlamydia trachomatis is an obligate intracellular bacterial pathogen and the second leading cause of sexually transmitted infections in the US. Infections cause significant morbidity and can lead to serious reproductive sequelae, including an epidemiological link to increased rates of reproductive cancers. One of the overt changes that infected cells exhibit is the development of genomic instability leading to multinucleation. Here we demonstrate that the induction of multinucleation is not conserved equally across chlamydial species; C. trachomatis L2 caused high levels of multinucleation, C. muridarum intermediate levels, and C. caviae had very modest effects on multinucleation. Our data show that at least two effector pathways together cause genomic instability during infection leading to multinucleation. We find that the highly conserved chlamydial protease CPAF is a key effector for one of these pathways. CPAF secretion is required for the loss of centrosome duplication regulation as well as inducing early mitotic exit. The second effector pathway involves the induction of centrosome position errors. This function is not conserved in three chlamydial species tested. Together these two pathways contribute to the induction of high levels of genomic instability and multinucleation seen in C. trachomatis infections.


Asunto(s)
Infecciones por Chlamydia/microbiología , Infecciones por Chlamydia/patología , Chlamydia trachomatis/fisiología , Células Gigantes/microbiología , Células Gigantes/patología , Transducción de Señal , Células 3T3 , Animales , Centrosoma , Segregación Cromosómica , ADN Bacteriano/metabolismo , Endopeptidasas/metabolismo , Técnica del Anticuerpo Fluorescente , Griseofulvina/farmacología , Células HeLa , Humanos , Ratones , Índice Mitótico , Modelos Biológicos , Mutación , Especificidad de la Especie , Huso Acromático/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...