Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 184
Filtrar
1.
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
3.
Sci Rep ; 11(1): 15745, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34344917

RESUMEN

In Europe, Ixodes ricinus is the most important vector of human infectious diseases, most notably Lyme borreliosis and tick-borne encephalitis virus. Multiple non-natural hosts of I. ricinus have shown to develop immunity after repeated tick bites. Tick immunity has also been shown to impair B. burgdorferi transmission. Most interestingly, multiple tick bites reduced the likelihood of contracting Lyme borreliosis in humans. A vaccine that mimics tick immunity could therefore potentially prevent Lyme borreliosis in humans. A yeast surface display library (YSD) of nymphal I. ricinus salivary gland genes expressed at 24, 48 and 72 h into tick feeding was constructed and probed with antibodies from humans repeatedly bitten by ticks, identifying twelve immunoreactive tick salivary gland proteins (TSGPs). From these, three proteins were selected for vaccination studies. An exploratory vaccination study in cattle showed an anti-tick effect when all three antigens were combined. However, immunization of rabbits did not provide equivalent levels of protection. Our results show that YSD is a powerful tool to identify immunodominant antigens in humans exposed to tick bites, yet vaccination with the three selected TSGPs did not provide protection in the present form. Future efforts will focus on exploring the biological functions of these proteins, consider alternative systems for recombinant protein generation and vaccination platforms and assess the potential of the other identified immunogenic TSGPs.


Asunto(s)
Antígenos/aislamiento & purificación , Ixodes/inmunología , Enfermedad de Lyme/transmisión , Glándulas Salivales/inmunología , Proteínas y Péptidos Salivales/inmunología , Mordeduras de Garrapatas/inmunología , Infestaciones por Garrapatas/inmunología , Animales , Antígenos/sangre , Antígenos/inmunología , Borrelia burgdorferi/aislamiento & purificación , Bovinos , Técnicas de Visualización de Superficie Celular/métodos , Femenino , Humanos , Inmunización , Enfermedad de Lyme/sangre , Enfermedad de Lyme/parasitología , Masculino , Fragmentos de Péptidos/inmunología , Biblioteca de Péptidos , Conejos , Saccharomyces cerevisiae , Infestaciones por Garrapatas/parasitología
4.
Front Immunol ; 12: 704621, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34322135

RESUMEN

The lack of tools for the precise manipulation of the tick microbiome is currently a major limitation to achieve mechanistic insights into the tick microbiome. Anti-tick microbiota vaccines targeting keystone bacteria of the tick microbiota alter tick feeding, but their impact on the taxonomic and functional profiles of the tick microbiome has not been tested. In this study, we immunized a vertebrate host model (Mus musculus) with live bacteria vaccines targeting keystone (i.e., Escherichia-Shigella) or non-keystone (i.e., Leuconostoc) taxa of tick microbiota and tested the impact of bacterial-specific antibodies (Abs) on the structure and function of tick microbiota. We also investigated the effect of these anti-microbiota vaccines on mice gut microbiota composition. Our results showed that the tick microbiota of ticks fed on Escherichia coli-immunized mice had reduced Escherichia-Shigella abundance and lower species diversity compared to ticks fed on control mice immunized with a mock vaccine. Immunization against keystone bacteria restructured the hierarchy of nodes in co-occurrence networks and reduced the resistance of the bacterial network to taxa removal. High levels of E. coli-specific IgM and IgG were negatively correlated with the abundance of Escherichia-Shigella in tick microbiota. These effects were not observed when Leuconostoc was targeted with vaccination against Leuconostoc mesenteroides. Prediction of functional pathways in the tick microbiome using PICRUSt2 revealed that E. coli vaccination reduced the abundance of lysine degradation pathway in tick microbiome, a result validated by qPCR. In contrast, the gut microbiome of immunized mice showed no significant alterations in the diversity, composition and abundance of bacterial taxa. Our results demonstrated that anti-tick microbiota vaccines are a safe, specific and an easy-to-use tool for manipulation of vector microbiome. These results guide interventions for the control of tick infestations and pathogen infection/transmission.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Bacterias , Vacunas Bacterianas , Microbioma Gastrointestinal/inmunología , Ixodes , Animales , Bacterias/clasificación , Bacterias/inmunología , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/farmacología , Ixodes/inmunología , Ixodes/microbiología , Ratones
5.
Parasit Vectors ; 14(1): 303, 2021 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-34090497

RESUMEN

BACKGROUND: An epitope, Galα1-3Galß1-4GlcNAc-R, termed α-gal, is present in glycoconjugates of New World monkeys (platyrrhines) and other mammals but not in hominoids and Old World monkeys (catarrhines). The difference is due to the inactivation of α1-3 galactosyl transferase (α1-3 GT) genes in catarrhines. Natural antibodies to α-gal are therefore developed in catarrhines but not platyrrhines and other mammals. Hypersensitivity reactions are commonly elicited by mosquito and tick vector bites. IgE antibodies against α-gal cause food allergy to red meat in persons who have been exposed to tick bites. Three enzymes synthesising the terminal α1-3-linked galactose in α-gal, that are homologous to mammalian α and ß1-4 GTs but not mammalian α1-3 GTs, were recently identified in the tick vector Ixodes scapularis. IgG and IgM antibodies to α-gal are reported to protect against malaria because mosquito-derived sporozoites of malaria parasites express α-gal on their surface. This article explores the possibility that the α-gal in sporozoites are acquired from glycoconjugates synthesised by mosquitoes rather than through de novo synthesis by sporozoites. METHODS: The presence of proteins homologous to the three identified tick α1-3 GTs and mammalian α1-3 GTs in two important mosquito vectors, Aedes aegypti and Anopheles gambiae, as well as Plasmodium malaria parasites, was investigated by BLASTp analysis to help clarify the source of the α-gal on sporozoite surfaces. RESULTS: Anopheles gambiae and Ae. aegypti possessed several different proteins homologous to the three I. scapularis proteins with α1-3 GT activity, but not mammalian α1-3 GTs. The putative mosquito α1-3 GTs possessed conserved protein domains characteristic of glycosyl transferases. However, the genus Plasmodium lacked proteins homologous to the three I. scapularis proteins with α1-3 GT activity and mammalian α1-3 GTs. CONCLUSIONS: The putative α1-3 GTs identified in the two mosquito vectors may synthesise glycoconjugates containing α-gal that can be transferred to sporozoite surfaces before they are inoculated into skin during blood feeding. The findings merit further investigation because of their implications for immunity against malaria, hypersensitivity to mosquito bites, primate evolution, and proposals for immunisation against α-gal.


Asunto(s)
Galactosiltransferasas/genética , Hipersensibilidad , Proteínas de Insectos/inmunología , Ixodes/química , Malaria/inmunología , Mosquitos Vectores/química , Mordeduras de Garrapatas/inmunología , Alérgenos/inmunología , Animales , Vectores de Enfermedades , Galactosiltransferasas/inmunología , Humanos , Hipersensibilidad/prevención & control , Inmunoglobulina E/inmunología , Proteínas de Insectos/genética , Ixodes/enzimología , Ixodes/genética , Ixodes/inmunología , Malaria/prevención & control , Mosquitos Vectores/enzimología , Mosquitos Vectores/genética
6.
FEBS Lett ; 595(12): 1622-1638, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33960414

RESUMEN

Ixodes scapularis ticks feed on humans and other vertebrate hosts and transmit several pathogens of public health concern. Tick saliva is a complex mixture of bioactive proteins, lipids and immunomodulators, such as I. scapularis sphingomyelinase (IsSMase)-like protein, an ortholog of dermonecrotoxin SMase D found in the venom of Loxosceles spp. of spiders. IsSMase modulates the host immune response towards Th2, which suppresses Th1-mediated cytokines to facilitate pathogen transmission. Arboviruses utilize exosomes for their transmission from tick to the vertebrate host, and exosomes derived from tick saliva/salivary glands suppress C-X-C motif chemokine ligand 12 and interleukin-8 immune response(s) in human skin to delay wound healing and repair processes. IsSMase affects also viral replication and exosome biogenesis, thereby inhibiting tick-to-vertebrate host transmission of pathogenic exosomes. In this review, we elaborate on exosomes and their biogenesis as potential candidates for developing novel control measure(s) to combat tick-borne diseases. Such targets could help with the development of an efficient anti-tick vaccine for preventing the transmission of tick-borne pathogens.


Asunto(s)
Infecciones por Arbovirus , Arbovirus/inmunología , Proteínas de Artrópodos/inmunología , Factores Inmunológicos/inmunología , Ixodes , Esfingomielina Fosfodiesterasa/inmunología , Animales , Infecciones por Arbovirus/inmunología , Infecciones por Arbovirus/prevención & control , Infecciones por Arbovirus/transmisión , Humanos , Ixodes/inmunología , Ixodes/virología , Glándulas Salivales/inmunología , Glándulas Salivales/virología , Células TH1/inmunología , Células Th2/inmunología
7.
Front Immunol ; 12: 615011, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33717102

RESUMEN

Introduction: Borrelia burgdorferi sensu lato (sl) is the causative agent of Lyme borreliosis. Currently there is no human vaccine against Lyme borreliosis, and most research focuses on recombinant protein vaccines. DNA tattoo vaccination with B. afzelii strain PKo OspC in mice has proven to be fully protective against B. afzelii syringe challenge and induces a favorable humoral immunity compared to recombinant protein vaccination. Alternatively, several recombinant protein vaccines based on tick proteins have shown promising effect in tick-bite infection models. In this study, we evaluated the efficacy of DNA vaccines against Borrelia OspC or tick antigens in a tick-bite infection model. Method: We vaccinated C3H/HeN mice with OspC using a codon-optimized DNA vaccine or with recombinant protein. We challenged these mice with B. burgdorferi sensu stricto (ss)-infected Ixodes scapularis nymphs. Subsequently, we vaccinated C3H/HeN mice with DNA vaccines coding for tick proteins for which recombinant protein vaccines have previously resulted in interference with tick feeding and/or Borrelia transmission: Salp15, tHRF, TSLPI, and Tix-5. These mice were also challenged with B. burgdorferi ss infected Ixodes scapularis nymphs. Results: DNA tattoo and recombinant OspC vaccination both induced total IgG responses. Borrelia cultures and DNA loads of skin and bladder remained negative in the mice vaccinated with OspC DNA vaccination, except for one culture. DNA vaccines against tick antigens Salp15 and Tix-5 induced IgG responses, while those against tHRF and TSLPI barely induced any IgG response. In addition, Borrelia cultures, and DNA loads from mice tattooed with DNA vaccines against tick proteins TSLPI, Salp15, tHRF, and Tix-5 were all positive. Conclusion: A DNA tattoo vaccine against OspC induced high specific IgG titers and provided near total protection against B. burgdorferi ss infection by tick challenge. In contrast, DNA tattoo vaccines against tick proteins TSLPI, Salp15, tHRF, and Tix-5 induced low to moderate IgG titers and did not provide protection. Therefore, DNA tattoo vaccination does not seem a suitable vaccine strategy to identify, or screen for, tick antigens for anti-tick vaccines. However, DNA tattoo vaccination is a straightforward and effective vaccination platform to assess novel B. burgdorferi sl antigen candidates in a relevant tick challenge model.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas de Artrópodos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Borrelia burgdorferi/inmunología , Ixodes/inmunología , Vacunas contra Enfermedad de Lyme/inmunología , Enfermedad de Lyme/prevención & control , Vacunas de ADN/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/genética , Proteínas de la Membrana Bacteriana Externa/genética , Borrelia burgdorferi/genética , Femenino , Inmunización , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Enfermedad de Lyme/transmisión , Ratones
8.
Sci Rep ; 11(1): 6095, 2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33731754

RESUMEN

Ixodes scapularis ticks transmit multiple pathogens, including Borrelia burgdorferi sensu stricto, and encode many proteins harboring epidermal growth factor (EGF)-like domains. We show that I. scapularis produces multiple orthologs for Bm86, a widely studied tick gut protein considered as a target of an anti-tick vaccine, herein termed as Is86. We show that Is86 antigens feature at least three identifiable regions harboring EGF-like domains (termed as EGF-1, EGF-2, and EGF-3) and are differentially upregulated during B. burgdorferi infection. Although the RNA interference-mediated knockdown of Is86 genes did not show any influences on tick engorgement or B. burgdorferi sensu stricto persistence, the immunization of murine hosts with specific recombinant EGF antigens marginally reduced spirochete loads in the skin, in addition to affecting tick blood meal engorgement and molting. However, given the borderline impact of EGF immunization on tick engorgement and pathogen survival in the vector, it is unlikely that these antigens, at least in their current forms, could be developed as potential vaccines. Further investigations of the biological significance of Is86 (and other tick antigens) would enrich our knowledge of the intricate biology of ticks, including their interactions with resident pathogens, and contribute to the development of anti-tick measures to combat tick-borne illnesses.


Asunto(s)
Anticuerpos/inmunología , Proteínas de Artrópodos/inmunología , Borrelia burgdorferi/inmunología , Conducta Alimentaria , Ixodes/inmunología , Enfermedad de Lyme/inmunología , Animales , Ratones
9.
Am J Trop Med Hyg ; 104(1): 175-183, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33258439

RESUMEN

In many regions where ticks negatively impact public health or economic production, multiple medically important tick species may have overlapping geographic distribution, and in North America, this includes members of Ixodes, Dermacentor, and Amblyomma genera. Acquired tick resistance is the process by which some animals develop an immune response against feeding ticks after one or more exposures. This form of immunity can restrict the ability of ticks to feed and may inhibit transmission of pathogens. Likewise, many proteins present in tick saliva are conserved among tick species, and prior studies have reported cross-protective host immunity against certain combinations of ticks. In this study, we used a guinea pig model to assess whether host resistance against Ixodes scapularis could confer protection against two other medically important tick vectors, Dermacentor variabilis and Amblyomma americanum. Tick challenges using nymphs were used to induce host resistance against a primary species, followed by additional challenge using a secondary tick species. Tick attachment to hosts and engorgement weights were reduced significantly for D. variabilis and A. americanum feeding on I. scapularis-sensitized hosts. Reciprocally, I. scapularis engorgement weights were reduced to a lesser extent, and attachment was unaffected when feeding on hosts sensitized with either D. variabilis or A. americanum. These results indicate that immunity against I. scapularis could potentially be exploited for use in an anti-tick vaccine targeting multiple tick species and their associated pathogens.


Asunto(s)
Vectores Arácnidos/inmunología , Susceptibilidad a Enfermedades/inmunología , Cobayas , Ixodes/inmunología , Enfermedades de los Roedores/parasitología , Infestaciones por Garrapatas/veterinaria , Animales , Ciencia de los Animales de Laboratorio , Enfermedades de los Roedores/inmunología , Infestaciones por Garrapatas/inmunología
10.
Parasite Immunol ; 43(5): e12816, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33368329

RESUMEN

The bacterial pathogen Borrelia burgdorferi is the causative agent of Lyme disease and is transmitted to humans through an Ixodes tick vector. B. burgdorferi is able to survive in both mammalian and tick hosts through careful modulation of its gene expression. This allows B. burgdorferi to adapt to the environmental and nutritional changes that occur when it is transmitted between the two hosts. Distinct interactions between the spirochete and its host occur at every step of the enzootic cycle and dictate the ability of the spirochete to survive until the next stage of the cycle. Studying the interface between B. burgdorferi, the Ixodes tick vector and the natural mammalian reservoirs has been made significantly more feasible through the complete genome sequences of the organisms and the advent of high throughput screening technologies. Ultimately, a thorough investigation of the interplay between the two domains (and two phyla within one domain) is necessary in order to completely understand how the pathogen is transmitted.


Asunto(s)
Vectores Arácnidos/microbiología , Borrelia burgdorferi/fisiología , Interacciones Microbiota-Huesped/fisiología , Ixodes/microbiología , Enfermedad de Lyme/microbiología , Mamíferos/microbiología , Animales , Vectores Arácnidos/inmunología , Borrelia burgdorferi/genética , Expresión Génica , Humanos , Ixodes/inmunología , Enfermedad de Lyme/epidemiología , Enfermedad de Lyme/transmisión , Mamíferos/sangre , Mamíferos/parasitología , Microbiota , Ninfa/microbiología , Glándulas Salivales/microbiología
11.
Trends Microbiol ; 28(11): 889-899, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32482556

RESUMEN

The complement system is an ancient arm of the innate immune system that plays important roles in pathogen recognition and elimination. Upon activation by microbes, complement opsonizes bacterial surfaces, recruits professional phagocytes, and causes bacteriolysis. Borreliella species are spirochetal bacteria that are transmitted to vertebrate hosts via infected Ixodes ticks and are the etiologic agents of Lyme disease. Pathogens that traffic in blood and other body fluids, like Borreliella, have evolved means to evade complement. Lyme disease spirochetes interfere with complement by producing a small arsenal of outer-surface lipoproteins that bind host complement components and manipulate their native activities. Here we review the current landscape of complement evasion by Lyme disease spirochetes and provide an update on recent discoveries.


Asunto(s)
Borrelia burgdorferi/inmunología , Proteínas del Sistema Complemento/inmunología , Evasión Inmune , Enfermedad de Lyme/inmunología , Enfermedad de Lyme/microbiología , Animales , Borrelia burgdorferi/genética , Borrelia burgdorferi/fisiología , Humanos , Inmunidad Innata , Ixodes/inmunología , Ixodes/microbiología , Ixodes/fisiología , Enfermedad de Lyme/transmisión
12.
Curr Protein Pept Sci ; 21(5): 517-526, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31613726

RESUMEN

Lyme disease (LD) is an infectious disease caused by the spirochetes of genus borrelia, which are transmitted by the ticks of the genus ixodes. LD is transmitted by the spirochete B. burgdorferi sensu lato. Once in contact with the host through a tick bite, the pathogen comes into contact with the host defense, and must escape this machinery to establish LD, thus using a large number of mechanisms involving the vector of the pathogen, the pathogen itself and also the host. The initial diagnosis of the disease can be made based on the clinical symptoms of LD and the disease can be treated and cured with antibiotics if the diagnosis is made early in the beginning of the disease. Contrariwise, if LD is left untreated, the pathogen disseminates throughout the tissues and organs of the body, where it establishes different types of disease manifestations. In the nervous system, the inflammation caused by B. burgdorferi is known as Lyme neuroborreliosis (LNB). LNB is one of the principal manifestations of LD. In this review, we systematically describe the different molecular interactions among B. burgdorferi, the vector (tick) and the mammalian host.


Asunto(s)
Vectores Arácnidos/microbiología , Proteínas Bacterianas/genética , Borrelia burgdorferi/patogenicidad , Interacciones Huésped-Patógeno/genética , Ixodes/microbiología , Enfermedad de Lyme/genética , Proteínas de la Membrana/genética , Receptores de Superficie Celular/genética , Animales , Vectores Arácnidos/inmunología , Proteínas Bacterianas/inmunología , Borrelia burgdorferi/inmunología , Citocinas/genética , Citocinas/inmunología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/inmunología , Humanos , Ixodes/inmunología , Enfermedad de Lyme/inmunología , Enfermedad de Lyme/microbiología , Enfermedad de Lyme/patología , Proteínas de la Membrana/inmunología , Sistema Nervioso/inmunología , Sistema Nervioso/microbiología , Sistema Nervioso/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Receptores de Superficie Celular/inmunología , Saliva/microbiología , Transducción de Señal
14.
Ticks Tick Borne Dis ; 10(6): 101269, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31445875

RESUMEN

Tick innate immunity involves humoral and cellular responses. Among the humoral effector molecules in ticks are the defensins which are a family of small peptides with a conserved γ-core motif that is crucial for their antimicrobial activity. Defensin families have been identified in several hard and soft tick species. However, little is known about the presence and antimicrobial activity of defensins from the Australian paralysis tick Ixodes holocyclus. In this study the I. holocyclus transcriptome was searched for the presence of defensins. Unique and non-redundant defensin sequences were identified and designated as holosins 1 - 5. The antimicrobial activity of holosins 2 and 3 and of the predicted γ-cores of holosins 1-4 (HoloTickCores 1-4), was assessed using Gram-negative and Gram-positive bacteria as well as the fungus Fusarium graminearum and the yeast Candida albicans. All holosins had molecular features that are conserved in other tick defensins. Furthermore holosins 2 and 3 were very active against the Gram-positive bacteria Staphylococcus aureus and Listeria grayi. Holosins 2 and 3 were also active against F. graminearum and C. albicans and 5 µM of peptide abrogate the growth of these microorganisms. The activity of the synthetic γ-cores was lower than that of the mature defensins apart from HoloTickCore 2 which had activity comparable to mature holosin 2 against the Gram-negative bacterium Escherichia coli. This study reveals the presence of a multigene defensin family in I. holocyclus with wide antimicrobial activity.


Asunto(s)
Antibacterianos/farmacología , Antifúngicos/farmacología , Defensinas/genética , Defensinas/inmunología , Ixodes/genética , Ixodes/inmunología , Secuencia de Aminoácidos , Animales , Antibacterianos/química , Antifúngicos/química , Proteínas de Artrópodos/química , Proteínas de Artrópodos/genética , Proteínas de Artrópodos/inmunología , Australia , Candida albicans/efectos de los fármacos , Defensinas/química , Fusarium/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Filogenia , Alineación de Secuencia , Transcriptoma
15.
Proc Natl Acad Sci U S A ; 116(1): 205-210, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30559180

RESUMEN

The E3 ubiquitin ligase X-linked inhibitor of apoptosis (XIAP) acts as a molecular rheostat for the immune deficiency (IMD) pathway of the tick Ixodes scapularis How XIAP activates the IMD pathway in response to microbial infection remains ill defined. Here, we identified the XIAP enzymatic substrate p47 as a positive regulator of the I. scapularis IMD network. XIAP polyubiquitylates p47 in a lysine 63-dependent manner and interacts with the p47 ubiquitin-like (UBX) module. p47 also binds to Kenny (IKKγ/NEMO), the regulatory subunit of the inhibitor of nuclear factor (NF)- κB kinase complex. Replacement of the amino acid lysine to arginine within the p47 linker region completely abrogated molecular interactions with Kenny. Furthermore, mitigation of p47 transcription levels through RNA interference in I. scapularis limited Kenny accumulation, reduced phosphorylation of IKKß (IRD5), and impaired cleavage of the NF-κB molecule Relish. Accordingly, disruption of p47 expression increased microbial colonization by the Lyme disease spirochete Borrelia burgdorferi and the rickettsial agent Anaplasma phagocytophilum Collectively, we highlight the importance of ticks for the elucidation of paradigms in arthropod immunology. Manipulating immune signaling cascades within I. scapularis may lead to innovative approaches to reducing the burden of tick-borne diseases.


Asunto(s)
Ixodes/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/fisiología , Anaplasma , Animales , Proteínas de Artrópodos/metabolismo , Proteínas de Artrópodos/fisiología , Borrelia burgdorferi , Drosophila , Técnicas de Inactivación de Genes , Ixodes/microbiología , Ixodes/fisiología , FN-kappa B/metabolismo , Dominios Proteicos , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/fisiología
17.
Artículo en Inglés | MEDLINE | ID: mdl-29896452

RESUMEN

Although Ixodes scapularis and other related tick species are considered prolific vectors for a number of important human diseases, many aspects of their biology, microbial interactions, and immunity are largely unknown; in particular, how these ancient vectors recognize invading pathogens like Borrelia burgdorferi and influence their persistence. The analysis of the Ixodes genome and a limited set of transcriptomic data have established that ticks encode many components of classical immune pathways; yet at the same time, they lack many key orthologs of these recognition networks. Therefore, whether a given immune pathway is active in Ixodes ticks and how precisely they exert its microbicidal functions are only incompletely delineated. A few recent studies have suggested that classical pathways like the Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) as well as immunodeficiency (IMD) pathways are fully functional in I. scapularis, and upon challenge with microbes, generate potent microbicidal responses against diverse tick-borne pathogens including B. burgdorferi. These studies also highlight novel concepts of vector immunity that include both a direct and an indirect mode of recognition of pathogens, as well as the influence of the gut microbiome, which ultimately dictates the outcome of a robust microbicidal response. Further understanding of how Ixodes ticks recognize and suppress invading microbes like B. burgdorferi will enrich our fundamental knowledge of vector immunobiology, thereby contributing to the development of future interventions to better control the tick-borne pathogen.


Asunto(s)
Borrelia burgdorferi/inmunología , Inmunidad , Ixodes/inmunología , Enfermedad de Lyme/inmunología , Animales , Antiinfecciosos , Vectores Arácnidos/genética , Vectores Arácnidos/inmunología , Vectores Arácnidos/microbiología , Borrelia burgdorferi/patogenicidad , Microbioma Gastrointestinal/inmunología , Genoma de los Insectos , Interacciones Huésped-Patógeno/inmunología , Ixodes/genética , Ixodes/microbiología , Enfermedad de Lyme/microbiología , Transducción de Señal , Transcriptoma , Tirosina/análogos & derivados , Tirosina/genética , Tirosina/inmunología , Tirosina/metabolismo
18.
Eur Ann Allergy Clin Immunol ; 50(1): 45-47, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29350022

RESUMEN

Summary: We report the case of a 38-year-old man who was bitten several times during his life by a tick. He didn't report any previous history of anaphylaxis after the ingestion of red meat. The serum specific IgE showed positivity to α-Gal. The proximity of the bits didn't increase the titer of IgE antibodies to alpha-gal. We could hypothesize that the frequency of the exposure to the tick Corresponding author bites and the amount of tick bites during his lifetime induced a sort of tolerance in this patient.


Asunto(s)
Alérgenos/inmunología , Anafilaxia/diagnóstico , Hipersensibilidad a los Alimentos/diagnóstico , Mordeduras de Garrapatas/diagnóstico , Enfermedades por Picaduras de Garrapatas/diagnóstico , alfa-Galactosidasa/inmunología , Adulto , Animales , Prueba de Desgranulación de los Basófilos , Humanos , Hipersensibilidad Tardía , Inmunoglobulina E/metabolismo , Italia , Ixodes/inmunología , Masculino , Proteínas de la Carne/inmunología , Carne Roja
19.
Dev Comp Immunol ; 79: 86-94, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29061482

RESUMEN

Ticks possess components of a primordial complement system that presumably play a role in the interaction of the tick immune system with tick-borne pathogens and affect their transmission. Here we characterized a novel complement component, tagged as IrC2/Bf, from the hard tick Ixodes ricinus, the principal vector of Lyme disease in Europe. IrC2/Bf is a multi-domain molecule composed of 5-7 CCP modules, varied by alternative splicing, followed by a von Willebrand factor A domain and a C-terminal trypsin-like domain. The primary structure and molecular architecture of IrC2/Bf displays the closest homology to the C3-complement component convertases described in horseshoe crabs. The irc2/bf gene is mainly expressed in the tick fat body associated with the trachea and, as determined by western blotting, the protein is present in low amounts in tick hemolymph. Expression of irc2/bf mRNA was significantly up-regulated in response to the intra-hemocoelic injection of the yeast Candida albicans and all tested Borrelia sp. strains (B. burgdorferi NE5264, B. burgdorferi CB26, B. garinii MSLB, B. afzelii CB43), but was not affected by injection of model Gram-negative and Gram-positive bacteria or the aseptic injection control. In-line with these results, RNAi-mediated silencing of irc2/bf inhibited phagocytosis of B. afzelii and C. albicans but not the other bacteria. Tissue expression profiles, specific responses to microbial challenges, and patterns of phagocytic phenotypes upon RNAi silencing observed for IrC2/Bf match well with the previously reported characteristics of I. ricinus C3-related molecule 1 (IrC3-1). Therefore we presume that IrC2/Bf functions as a convertase in the same complement activation pathway protecting ticks against yeast and Borrelia infection.


Asunto(s)
Borrelia burgdorferi/inmunología , Candida albicans/inmunología , Candidiasis/inmunología , Complemento C3/metabolismo , Hemocitos/inmunología , Proteínas de Insectos/metabolismo , Ixodes/inmunología , Enfermedad de Lyme/inmunología , Animales , Activación de Complemento , Complemento C3/genética , Vectores de Enfermedades , Hemocitos/microbiología , Interacciones Huésped-Patógeno , Humanos , Proteínas de Insectos/genética , Fagocitosis , ARN Interferente Pequeño/genética
20.
Artículo en Inglés | MEDLINE | ID: mdl-29250492

RESUMEN

Emerging and re-emerging diseases transmitted by blood feeding arthropods are significant global public health problems. Ticks transmit the greatest variety of pathogenic microorganisms of any blood feeding arthropod. Infectious agents transmitted by ticks are delivered to the vertebrate host together with saliva at the bite site. Tick salivary glands produce complex cocktails of bioactive molecules that facilitate blood feeding and pathogen transmission by modulating host hemostasis, pain/itch responses, wound healing, and both innate and adaptive immunity. In this study, we utilized Illumina Next Generation Sequencing to characterize the transcriptional immunoprofile of cutaneous immune responses to Ixodes ricinus transmitted tick-borne encephalitis virus (TBEV). A comparative immune gene expression analysis of TBEV-infected and uninfected tick feeding sites was performed. Our analysis reveals that ticks create an inflammatory environment at the bite site during the first 3 h of feeding, and significant differences in host responses were observed between TBEV-infected and uninfected tick feeding. Gene-expression analysis reveals modulation of inflammatory genes after 1 and 3 h of TBEV-infected tick feeding. Transcriptional levels of genes specific to chemokines and cytokines indicated a neutrophil-dominated immune response. Immunohistochemistry of the tick feeding site revealed that mononuclear phagocytes and fibroblasts are the primary target cells for TBEV infection and did not detect TBEV antigens in neutrophils. Together, the transcriptional and immunohistochemistry results suggest that early cutaneous host responses to TBEV-infected tick feeding are more inflammatory than expected and highlight the importance of inflammatory chemokine and cytokine pathways in tick-borne flavivirus transmission.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/transmisión , Expresión Génica/inmunología , Interacciones Huésped-Parásitos/inmunología , Ixodes/inmunología , Animales , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Femenino , Flavivirus/inmunología , Flavivirus/patogenicidad , Secuenciación de Nucleótidos de Alto Rendimiento , Antígenos de Histocompatibilidad Clase II/inmunología , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Neutrófilos/inmunología , Piel/inmunología , Piel/patología , Mordeduras de Garrapatas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...