Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-38746284

RESUMEN

Ixodes scapularis ticks are an important vector for at least six tick-borne human pathogens, including the predominant North American Lyme disease spirochete Borrelia burgdorferi . The ability for these ticks to survive in nature is credited, in part, to their ability to feed on a variety of hosts without excessive activation of the proinflammatory branch of the vertebrate immune system. While the ability for nymphal ticks to feed on a variety of hosts has been well-documented, the host-parasite interactions between larval I. scapularis and different vertebrate hosts is relatively unexplored. Here we report on the changes in the vertebrate transcriptome present at the larval tick bite site using the natural I. scapularis host Peromyscus leucopus deermouse, a non-natural rodent host Mus musculus (BALB/c), and humans. We note substantially less evidence of activation of canonical proinflammatory pathways in P. leucopus compared to BALB/c mice and pronounced evidence of inflammation in humans. Pathway enrichment analyses revealed a particularly strong signature of interferon gamma, tumor necrosis factor, and interleukin 1 signaling at the BALB/c and human tick bite site. We also note that bite sites on BALB/c mice and humans, but not deermice, show activation of wound-healing pathways. These data provide molecular evidence of the coevolution between larval I. scapularis and P. leucopus as well as expand our overall understanding of I. scapularis feeding. Significance: Ixodes scapularis tick bites expose humans to numerous diseases in North America. While larval tick feeding enables pathogens to enter the tick population and eventually spread to humans, how larval ticks interact with mammals has been understudied compared to other tick stages. Here we examined the transcriptomic response of a natural I. scapularis rodent host ( Peromyscus leucopus ), a non-native I. scapularis rodent host ( Mus musculus ), and an incidental host (humans). We find that there are differences in how all three species respond to larval I. scapularis , with the natural host producing the smallest transcriptomic signature of a canonical proinflammatory immune response and the incidental human host producing the most robust signature of inflammation in response to the larval tick. These data expand our understanding of the pressures on ticks in the wild and inform our ability to model these interactions in laboratory settings.

2.
Artículo en Inglés | MEDLINE | ID: mdl-38606926

RESUMEN

AIMS: Aortic valve calcification (AVC) of surgical valve bioprostheses (BP) has been poorly explored. We aimed to evaluate in-vivo and ex-vivo BP AVC and its prognosis value. METHODS AND RESULTS: Between 2011 and 2019, AVC was assessed using in-vivo computed tomography (CT) in 361 patients who had undergone surgical valve replacement 6.4±4.3 years earlier. Ex-vivo CT scans were performed for 37 explanted BP. The in-vivo CT scans were interpretable for 342 patients (19 patients [5.2%], were excluded). These patients were 77.2±9.1 years old and 64.3% were male. Mean in-vivo AVC was 307±500 Agatston unit (AU). The AVC was 562±570 AU for the 183 (53.5%) patients with structural valve degeneration (SVD) and 13±43 AU for those without SVD (p<0.0001). In-vivo and ex-vivo AVC were strongly correlated (r=0.88, p<0.0001). An in-vivo AVC>100 AU (n=147, 43%) had a specificity of 96% for diagnosing Stage 2-3 SVD (area under the curve=0.92). Patients with AVC>100 AU had a worse outcome compared with those with AVC≤100 AU (n=195). In multivariable analysis, AVC was a predictor of overall mortality (hazard ratio [HR] and 95% confidence interval=1.16[1.04-1.29]; p=0.006), cardiovascular mortality (HR=1.22[1.04-1.43]; p=0.013), cardiovascular events (HR=1.28 [1.16-1.41]; p<0.0001), and re-intervention (HR=1.15 [1.06-1.25]; p<0.0001). After adjustment for Stage 2-3 SVD diagnosis, AVC remained a predictor of overall mortality (HR=1.20 [1.04-1.39]; p=0.015) and cardiovascular events (HR=1.25 [1.09-1.43]; p=0.001). CONCLUSION: CT scan is a reliable tool to assess BP leaflet calcification. An AVC>100 AU is tightly associated with SVD and it is a strong predictor of overall mortality and cardiovascular events.

3.
PLoS Pathog ; 19(7): e1011479, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37428812

RESUMEN

Buruli ulcer is a chronic infectious disease caused by Mycobacterium ulcerans. The pathogen persistence in host skin is associated with the development of ulcerative and necrotic lesions leading to permanent disabilities in most patients. However, few of diagnosed cases are thought to resolve through an unknown self-healing process. Using in vitro and in vivo mouse models and M. ulcerans purified vesicles and mycolactone, we showed that the development of an innate immune tolerance was only specific to macrophages from mice able to heal spontaneously. This tolerance mechanism depends on a type I interferon response and can be induced by interferon beta. A type I interferon signature was further detected during in vivo infection in mice as well as in skin samples from patients under antibiotics regiment. Our results indicate that type I interferon-related genes expressed in macrophages may promote tolerance and healing during infection with skin damaging pathogen.


Asunto(s)
Úlcera de Buruli , Interferón Tipo I , Mycobacterium ulcerans , Ratones , Animales , Úlcera de Buruli/microbiología , Macrófagos , Macrólidos , Tolerancia Inmunológica
4.
Science ; 379(6628): eabl3837, 2023 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-36634189

RESUMEN

Ancestral signaling pathways serve critical roles in metazoan development, physiology, and immunity. We report an evolutionary interspecies communication pathway involving a central Ixodes scapularis tick receptor termed Dome1, which acquired a mammalian cytokine receptor motif exhibiting high affinity for interferon-gamma (IFN-γ). Host-derived IFN-γ facilitates Dome1-mediated activation of the Ixodes JAK-STAT pathway. This accelerates tick blood meal acquisition and development while upregulating antimicrobial components. The Dome1-JAK-STAT pathway, which exists in most Ixodid tick genomes, regulates the regeneration and proliferation of gut cells-including stem cells-and dictates metamorphosis through the Hedgehog and Notch-Delta networks, ultimately affecting Ixodes vectorial competence. We highlight the evolutionary dependence of I. scapularis on mammalian hosts through cross-species signaling mechanisms that dually influence arthropod immunity and development.


Asunto(s)
Vectores Arácnidos , Interacciones Huésped-Parásitos , Ixodes , Quinasas Janus , Receptores de Citocinas , Factores de Transcripción STAT , Animales , Interferón gamma/metabolismo , Ixodes/genética , Ixodes/inmunología , Quinasas Janus/genética , Quinasas Janus/metabolismo , Transducción de Señal , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Interacciones Huésped-Parásitos/inmunología , Receptores de Citocinas/metabolismo , Vectores Arácnidos/inmunología
5.
Eur J Clin Microbiol Infect Dis ; 40(9): 2023-2028, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33686557

RESUMEN

During an epidemic period, we compared patients hospitalized for initial suspicion of COVID-19 but for whom an alternative diagnosis was finally retained (n = 152) with those who had COVID-19 (n = 222). Most common diagnoses were another infectious disease and heart failure. COVID-19-negative patients were more often active smokers had less often cough, fever, and digestive symptoms, as compared to the 222 COVID-19-positive patients. They had higher median neutrophil and lymphocyte counts and lower CRP level. In multivariate analysis, no current smoking, neurocognitive disorder, myalgia, and fibrinogen ≥4g/L were independently associated with a final diagnosis of COVID-19.


Asunto(s)
COVID-19/diagnóstico , Adulto , Anciano , COVID-19/terapia , COVID-19/virología , Hospitalización , Humanos , Masculino , Pacientes/estadística & datos numéricos , Estudios Retrospectivos , SARS-CoV-2/fisiología
7.
J Immunol ; 205(12): 3383-3389, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33168577

RESUMEN

Borrelia burgdorferi, the causative agent of Lyme disease, is transmitted by the bite of an infected tick. Once inoculated into the host dermis, it disseminates to various organs including distant skin sites, the heart, the joint and the nervous system. Most humans will develop an early skin manifestation called erythema migrans at the tick bite site. This can be followed by symptoms such as carditis, neuritis, meningitis, or arthritis if not treated. A specific mouse strain, C3H/HeN develops arthritis with B. burgdorferi infection whereas another strain, C57BL/6, develops minimal to no arthritis. Neither strain of mice show any skin signs of rash or inflammation. Factors that determine the presence of skin inflammation and the joint arthritis susceptibility in the host are only partially characterized. We show in this study that murine fibroblast-like synoviocytes display trained immunity, a program in some cells that results in increased inflammatory responses if the cell has previously come in contact with a stimulus, and that trained immunity in fibroblast-like synoviocytes tested ex vivo correlates with Lyme arthritis susceptibility. Conversely, skin fibroblasts do not exhibit trained immunity, which correlates with the absence of skin symptoms in these mice. Moreover, we demonstrate that the trained phenotype in FLS is affected by the cell environment, which depends on the host genetic background. Future studies expanding this initial report of the role of trained immunity on symptoms of B. burgdorferi infection may provide insight into the pathogenesis of disease in murine models.


Asunto(s)
Artritis/inmunología , Borrelia burgdorferi/inmunología , Inmunidad Innata , Memoria Inmunológica , Enfermedad de Lyme/inmunología , Sinoviocitos/inmunología , Animales , Artritis/genética , Artritis/patología , Femenino , Inflamación/inmunología , Inflamación/patología , Enfermedad de Lyme/genética , Enfermedad de Lyme/patología , Ratones , Ratones Noqueados , Sinoviocitos/patología
8.
Trends Parasitol ; 36(3): 304-314, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32007396

RESUMEN

The skin plays a key role in vector-borne diseases because it is the site where the arthropod coinoculates pathogens and its saliva. Lyme borreliosis, particularly well investigated in this context, is a multisystemic infectious disease caused by Borrelia burgdorferi sensu lato and transmitted by the hard tick Ixodes. Numerous in vitro studies were conducted to better understand the role of specific skin cells and tick saliva in host defense, vector feeding, and pathogen transmission. The skin was also evidenced in various animal models as the site of bacterial multiplication and persistence. We present the achievements in this field as well as the gaps that impede comprehensive knowledge of the disease pathophysiology and the development of efficient diagnostic tools and vaccines in humans.


Asunto(s)
Borrelia/fisiología , Enfermedad de Lyme/microbiología , Piel/microbiología , Animales , Borrelia/inmunología , Humanos , Ixodes/microbiología , Enfermedad de Lyme/diagnóstico , Enfermedad de Lyme/inmunología , Enfermedad de Lyme/prevención & control , Piel/inmunología
9.
J Infect Dis ; 221(9): 1438-1447, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-31758693

RESUMEN

Borrelia burgdorferi conserved gene products BB0406 and BB0405, members of a common B. burgdorferi paralogous gene family, share 59% similarity. Although both gene products can function as potential porins, only BB0405 is essential for infection. Here we show that, despite sequence homology and coexpression from the same operon, both proteins differ in their membrane localization attributes, antibody accessibility, and immunogenicity in mice. BB0406 is required for spirochete survival in mammalian hosts, particularly for the disseminated infection in distant organs. We identified that BB0406 interacts with laminin, one of the major constituents of the vascular basement membrane, and facilitates spirochete transmigration across host endothelial cell barriers. A better understanding of how B. burgdorferi transmigrates through dermal and tissue vascular barriers and establishes disseminated infections will contribute to the development of novel therapeutics to combat early infection.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Células Endoteliales/microbiología , Interacciones Huésped-Patógeno , Laminina/metabolismo , Enfermedad de Lyme/microbiología , Animales , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/genética , Borrelia burgdorferi/efectos de los fármacos , Borrelia burgdorferi/genética , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Expresión Génica , Marcación de Gen , Prueba de Complementación Genética , Humanos , Ratones , Ratones Endogámicos C3H , Mutación , Unión Proteica
10.
Ticks Tick Borne Dis ; 10(2): 433-441, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30595500

RESUMEN

Understanding the mechanism of pathogen transmission is essential for the development of strategies to reduce arthropod-borne diseases. The pharmaco- and immunomodulatory properties of insect and acarine saliva play an essential role in the efficiency of pathogen transmission. The skin as the site where arthropod saliva and pathogens are inoculated - represents the key interface in vector-borne diseases. We identified tick molecules potentially involved in pathogen transmission, using micro-HPLC and mass spectrometry, followed by in vitro assays on human skin cells. Histone H4 isolated from Ixodes ricinus salivary gland extract was identified as a molecule with a dissociating effect on human primary fibroblasts. This histone might be involved in the formation of the feeding pool formed around the tick mouthparts and responsible of tissue necrosis in the vertebrate host. Thanks to its selective antimicrobial activity, it may also sterilize the feeding pool and facilitate transmission of pathogens such as Borrelia burgdorferi sensu lato.


Asunto(s)
Fibroblastos/efectos de los fármacos , Ixodes/química , Enfermedad de Lyme/transmisión , Glándulas Salivales/química , Extractos de Tejidos/farmacología , Animales , Borrelia burgdorferi , Células Cultivadas , Cromatografía Líquida de Alta Presión , Femenino , Histonas/farmacología , Humanos , Enfermedad de Lyme/microbiología , Espectrometría de Masas , Extractos de Tejidos/química
11.
Cell Microbiol ; 21(2): e12885, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29934966

RESUMEN

Borrelia burgdorferi is the causative agent of Lyme disease that persists in a complex enzootic life cycle, involving Ixodes ticks and vertebrate hosts. The microbe invades ticks and vertebrate hosts in spite of active immune surveillance and potent microbicidal responses, and establishes long-term infection utilising mechanisms that are yet to be unravelled. The pathogen can cause multi-system disorders when transmitted to susceptible mammalian hosts, including in humans. In the past decades, several studies identified a limited number of B. burgdorferi gene-products critical for pathogen persistence, transmission between the vectors and the host, and host-pathogen interactions. This review will focus on the interactions between B. burgdorferi proteins, as well as between microbial proteins and host components, protein and non-protein components, highlighting their roles in pathogen persistence in the mammalian host. A better understanding of the contributions of protein interactions in the microbial virulence and persistence of B. burgdorferi would support development of novel therapeutics against the infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Borrelia burgdorferi/patogenicidad , Interacciones Huésped-Patógeno/fisiología , Enfermedad de Lyme/patología , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/genética , Borrelia burgdorferi/efectos de los fármacos , Borrelia burgdorferi/genética , Humanos , Ixodes/microbiología , Enfermedad de Lyme/tratamiento farmacológico , Enfermedad de Lyme/microbiología , Unión Proteica/fisiología , Mapas de Interacción de Proteínas , Virulencia , Factores de Virulencia/genética
12.
Proc Natl Acad Sci U S A ; 115(16): E3788-E3797, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29610317

RESUMEN

Borrelia burgdorferi is one of the few extracellular pathogens capable of establishing persistent infection in mammals. The mechanisms that sustain long-term survival of this bacterium are largely unknown. Here we report a unique innate immune evasion strategy of B. burgdorferi, orchestrated by a surface protein annotated as BBA57, through its modulation of multiple spirochete virulent determinants. BBA57 function is critical for early infection but largely redundant for later stages of spirochetal persistence, either in mammals or in ticks. The protein influences host IFN responses as well as suppresses multiple host microbicidal activities involving serum complement, neutrophils, and antimicrobial peptides. We also discovered a remarkable plasticity in BBA57-mediated spirochete immune evasion strategy because its loss, although resulting in near clearance of pathogens at the inoculum site, triggers nonheritable adaptive changes that exclude detectable nucleotide alterations in the genome but incorporate transcriptional reprograming events. Understanding the malleability in spirochetal immune evasion mechanisms that ensures their host persistence is critical for the development of novel therapeutic and preventive approaches to combat long-term infections like Lyme borreliosis.


Asunto(s)
Proteínas Bacterianas/fisiología , Borrelia burgdorferi/inmunología , Evasión Inmune , Lipoproteínas/fisiología , Proteínas de la Membrana/fisiología , Animales , Antígenos Bacterianos/inmunología , Péptidos Catiónicos Antimicrobianos/biosíntesis , Péptidos Catiónicos Antimicrobianos/genética , Vectores Arácnidos/microbiología , Proteínas Bacterianas/genética , Borrelia burgdorferi/genética , Borrelia burgdorferi/patogenicidad , Células Cultivadas , Proteínas del Sistema Complemento/inmunología , Citocinas/biosíntesis , Citocinas/genética , Femenino , Regulación Bacteriana de la Expresión Génica , Humanos , Ixodes/microbiología , Lipoproteínas/genética , Enfermedad de Lyme/inmunología , Enfermedad de Lyme/microbiología , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones SCID , Organismos Libres de Patógenos Específicos , Virulencia
13.
Sci Rep ; 8(1): 1910, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29382879

RESUMEN

In its natural infection cycle, the pathogen of Lyme borreliosis transits between a tick vector and a mammalian host. As relatively a minor fraction of spirochetes transits between the host and the vector precluding their reliable detection at early infection, artificial membrane feeders emerged as useful tools to study roles of spirochete proteins in pathogen entry, persistence, and exit through ticks. Here we report the development of a modified membrane feeder to study the role of a Borrelia burgdorferi surface protein called Lmp1 in spirochete transitions between the murine host and ticks. We show that our membrane feeder supports the blood meal engorgement process where ticks can acquire spirochetes from the feeder containing extremely low levels of pathogens (102 cells/ml of blood). Our data revealed that in comparison to wild-type spirochetes, lmp1 deletion mutants are significantly impaired for acquisition in naïve ticks as well as transmission from infected ticks. Taking together, our data suggest that Lmp1 plays an essential role in spirochete transitions between hosts and the vector. These studies also underscore the usefulness of artificial membrane feeding system as a valuable tool to study the role of B. burgdorferi gene-products in pathogen persistence in and passage through vector ticks.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Borrelia burgdorferi/metabolismo , Borrelia burgdorferi/patogenicidad , Enfermedad de Lyme/metabolismo , Enfermedad de Lyme/microbiología , Membranas/metabolismo , Garrapatas/metabolismo , Animales , Vectores de Enfermedades , Femenino , Interacciones Huésped-Patógeno/fisiología , Membranas Artificiales , Ratones , Ratones Endogámicos C3H
14.
Methods Mol Biol ; 1690: 319-327, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29032555

RESUMEN

The skin plays an essential role in the transmission of Lyme borreliosis since it is the first interface between the Ixodes tick and the host during the inoculation of Borrelia burgdorferi sensu lato. A better understanding of the inflammatory reaction at this location is key to develop better strategies (e.g., vaccine and diagnosis) to fight this disease. In vitro cell culture of resident skin cells might constitute an approach to decipher the complex interplay between the tick, the pathogen, and the vertebrate host.


Asunto(s)
Borrelia burgdorferi/fisiología , Técnicas de Cultivo de Célula/métodos , Dermatitis/patología , Fibroblastos/patología , Queratinocitos/patología , Enfermedad de Lyme/patología , Piel/patología , Animales , Vectores Arácnidos/microbiología , Vectores Arácnidos/fisiología , Borrelia burgdorferi/aislamiento & purificación , Células Cultivadas , Dermatitis/microbiología , Femenino , Fibroblastos/microbiología , Interacciones Huésped-Patógeno , Humanos , Ixodes/microbiología , Ixodes/fisiología , Queratinocitos/microbiología , Enfermedad de Lyme/microbiología , Piel/microbiología , Adulto Joven
15.
Parasit Vectors ; 10(1): 313, 2017 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-28655322

RESUMEN

BACKGROUND: Borrelia burgdorferi (sensu lato), the causative agent of Lyme borreliosis is a bacterium transmitted by hard ticks, Ixodes spp. Bacteria are injected into the host skin during the tick blood meal with tick saliva. There, Borrelia and saliva interact together with skin cells such as keratinocytes, fibroblasts, mast cells and other specific immune cells before disseminating to target organs. METHODS: To study the role of mast cells in the transmission of Lyme borreliosis, we isolated mouse primary mast cells from bone marrow and incubated them in the presence of Borrelia burgdorferi (sensu stricto) and tick salivary gland extract. We further analyzed their potential role in vivo, in a mouse model of deficient in mast cells (Kit wsh-/- mice). RESULTS: To our knowledge, we report here for the first time the bacteria ability to induce the inflammatory response of mouse primary mast cells. We show that OspC, a major surface lipoprotein involved in the early transmission of Borrelia, induces the degranulation of primary mast cells but has a limited effect on the overall inflammatory response of these cells. In contrast, whole bacteria have an opposite effect. We also show that mast cell activation is significantly inhibited by tick salivary gland extract. Finally, we demonstrate that mast cells are likely not the only host cells involved in the early transmission and dissemination of Borrelia since the use of mast cell deficient Kit wsh-/- mice shows a limited impact on these two processes in the context of this mouse genetic background. CONCLUSIONS: The absence of mast cells did not change the replication rate of Borrelia in the skin. However, in the absence of mast cells, Borrelia dissemination to the joints was faster. Mast cells do not control skin bacterial proliferation during primary infection and the establishment of the primary infection, as shown in the C57BL/6 mouse model studied. Nevertheless, the Borrelia induced cytotokine modulation on mast cells might be involved in long term and/or repeated infections and protect from Lyme borreliosis due to the development of a hypersensitivity to tick saliva.


Asunto(s)
Antígenos Bacterianos/metabolismo , Vectores Arácnidos/inmunología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Borrelia burgdorferi/inmunología , Ixodes/inmunología , Enfermedad de Lyme/inmunología , Mastocitos/inmunología , Animales , Antígenos Bacterianos/genética , Vectores Arácnidos/microbiología , Proteínas de la Membrana Bacteriana Externa/genética , Borrelia burgdorferi/fisiología , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Ixodes/microbiología , Enfermedad de Lyme/microbiología , Enfermedad de Lyme/transmisión , Mastocitos/microbiología , Ratones , Saliva/inmunología , Saliva/microbiología , Glándulas Salivales/inmunología , Glándulas Salivales/microbiología
16.
Exp Dermatol ; 25(1): 26-31, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26307945

RESUMEN

Ixodes hard tick induces skin injury by its sophisticated biting process. Its saliva plays a key role to enable an efficient blood meal that lasts for several days. We hypothesized that this feeding process may also be exploited by pathogens to facilitate their transmission, especially in the context of arthropod-borne diseases. To test this, we used Lyme borreliosis as a model. This bacterial infection is caused by Borrelia burgdorferi sensu lato transmitted by Ixodes. We co-incubated Borrelia with human keratinocytes in the presence of poly (I: C), a dsRNA TLR3 agonist generated by skin injury. This induced a strong cytokine response from human primary keratinocytes that was much greater than that induced by Borrelia alone. OspC, a TLR2/1 agonist and a major surface lipoprotein of Borrelia also amplified the process. Interestingly, tick saliva inhibited cytokine responses by keratinocytes to these TLR agonists. We propose that Borrelia uses the immunoprivileged site produced by tick saliva to facilitate its transmission.


Asunto(s)
Borrelia burgdorferi , Ixodes , Saliva/microbiología , Enfermedades de la Piel/microbiología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 3/metabolismo , Animales , Antígenos Bacterianos/metabolismo , Apoptosis , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proliferación Celular , Células Cultivadas , Citocinas/metabolismo , Humanos , Inflamación , Queratinocitos/citología , Queratinocitos/metabolismo , Queratinocitos/microbiología , Ligandos , Enfermedad de Lyme/inmunología , Ratones , Ratones Endogámicos C57BL , Poli I-C/química , ARN Bicatenario/química , Piel/inmunología , Piel/lesiones , Piel/microbiología , Enfermedades de la Piel/inmunología
17.
PLoS One ; 10(7): e0133195, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26197047

RESUMEN

Lyme disease is a multisystemic disorder caused by B. burgdorferi sl. The molecular basis for specific organ involvement is poorly understood. The skin plays a central role in the development of Lyme disease as the entry site of B. burgdorferi in which specific clones are selected before dissemination. We compared the skin inflammatory response (antimicrobial peptides, cytokines and chemokines) elicited by spirochete populations recovered from patients presenting different clinical manifestations. Remarkably, these spirochete populations induced different inflammatory profiles in the skin of C3H/HeN mice. As spirochete population transmitted into the host skin is heterogeneous, we isolated one bacterial clone from a population recovered from a patient with neuroborreliosis and compared its virulence to the parental population. This clone elicited a strong cutaneous inflammatory response characterized by MCP-1, IL-6 and antimicrobial peptides induction. Mass spectrometry of this clone revealed 110 overexpressed proteins when compared with the parental population. We further focused on the expression of nine bacterial surface proteins. bb0347 coding for a protein that interacts with host fibronectin, allowing bacterial adhesion to vascular endothelium and extracellular matrix, was found to be induced in host skin with another gene bb0213 coding for a hypothetical protein. These findings demonstrate the heterogeneity of the B. burgdorferi ss population and the complexity of the interaction involved early in the skin.


Asunto(s)
Borrelia burgdorferi/genética , Heterogeneidad Genética , Piel/microbiología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Borrelia burgdorferi/patogenicidad , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Fibronectinas/metabolismo , Flagelina/genética , Flagelina/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Ratones , Microbiota , Piel/metabolismo
18.
Eur J Dermatol ; 25 Suppl 1: 18-22, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26083670

RESUMEN

In the last years, the skin has been described as a major interface in arthropod borne diseases. Although it constitutes an efficient immune and physical barrier, pathogens have developed effective strategies to thwart the host. In this process, the arthropod plays a major role. For mosquitoes, the quick blood meal is made through an efficient inoculation process directly into the blood vessel. For the long lasting blood meal of hard ticks, the sophisticated biting pieces and the tick saliva provide potent tools to help pathogen transmission. Lyme borreliosis and leishmaniases have been particularly well investigated in this context.


Asunto(s)
Vectores Artrópodos/fisiología , Transmisión de Enfermedad Infecciosa , Interacciones Huésped-Patógeno , Fenómenos Fisiológicos de la Piel , Animales , Dengue/transmisión , Humanos , Leishmaniasis/transmisión , Malaria/transmisión , Enfermedades por Picaduras de Garrapatas/transmisión
19.
J Invest Dermatol ; 134(5): 1211-1219, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24552683

RESUMEN

The skin is a critical barrier between hosts and pathogens in arthropod-borne diseases. It harbors many resident cells and specific immune cells to arrest or limit infections by secreting inflammatory molecules or by directly killing pathogens. However, some pathogens are able to use specific skin cells and arthropod saliva for their initial development, to hide from the host immune system, and to establish persistent infection in the vertebrate host. A better understanding of the initial mechanisms taking place in the skin should allow the development of new strategies to fight these vector-borne pathogens that are spread worldwide and are of major medical importance.


Asunto(s)
Vectores Artrópodos , Artrópodos/inmunología , Sistema Inmunológico , Infecciones/transmisión , Piel/inmunología , Animales , Vectores Artrópodos/microbiología , Vectores Artrópodos/parasitología , Vectores Artrópodos/virología , Humanos , Sistema Inmunológico/microbiología , Sistema Inmunológico/parasitología , Sistema Inmunológico/virología , Infecciones/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA