Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(20): e2208673120, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-37155900

RESUMEN

The immune deficiency (IMD) pathway directs host defense in arthropods upon bacterial infection. In Pancrustacea, peptidoglycan recognition proteins sense microbial moieties and initiate nuclear factor-κB-driven immune responses. Proteins that elicit the IMD pathway in non-insect arthropods remain elusive. Here, we show that an Ixodes scapularis homolog of croquemort (Crq), a CD36-like protein, promotes activation of the tick IMD pathway. Crq exhibits plasma membrane localization and binds the lipid agonist 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoglycerol. Crq regulates the IMD and jun N-terminal kinase signaling cascades and limits the acquisition of the Lyme disease spirochete B. burgdorferi. Additionally, nymphs silenced for crq display impaired feeding and delayed molting to adulthood due to a deficiency in ecdysteroid synthesis. Collectively, we establish a distinct mechanism for arthropod immunity outside of insects and crustaceans.


Asunto(s)
Artrópodos , Infecciones Bacterianas , Borrelia burgdorferi , Ixodes , Enfermedad de Lyme , Animales , Ixodes/microbiología , Borrelia burgdorferi/genética , FN-kappa B , Enfermedad de Lyme/microbiología
2.
J Infect Dis ; 225(1): 135-145, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-34139755

RESUMEN

Human babesiosis is an emerging tick-borne malaria-like illness caused by Babesia parasites following their development in erythrocytes. Here, we show that a mutation in the Babesia microti mitochondrial cytochrome b (Cytb) that confers resistance to the antibabesial drug ELQ-502 decreases parasite fitness in the arthropod vector. Interestingly, whereas the mutant allele does not affect B. microti fitness during the mammalian blood phase of the parasite life cycle and is genetically stable as parasite burden increases, ELQ-502-resistant mutant parasites developing in the tick vector are genetically unstable with a high rate of the wild-type allele emerging during the nymphal stage. Furthermore, we show that B. microti parasites with this mutation are transmitted from the tick to the host, raising the possibility that the frequency of Cytb resistance mutations may be decreased by passage through the tick vector, but could persist in the environment if present when ticks feed.


Asunto(s)
Antiprotozoarios/farmacología , Babesia/genética , Babesiosis/tratamiento farmacológico , Babesiosis/transmisión , Citocromos b/genética , Resistencia a Medicamentos/genética , Ixodes , Quinolonas/farmacología , Garrapatas , Animales , Babesia/efectos de los fármacos , Babesia/crecimiento & desarrollo , Babesiosis/diagnóstico , Citocromos b/efectos de los fármacos , Eritrocitos/parasitología , Humanos , Mutación , Parásitos
3.
Parasite Immunol ; 43(5): e12808, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33187012

RESUMEN

Acquired tick resistance is a phenomenon wherein the host elicits an immune response against tick salivary components upon repeated tick infestations. The immune responses, potentially directed against critical salivary components, thwart tick feeding, and the animal becomes resistant to subsequent tick infestations. The development of tick resistance is frequently observed when ticks feed on non-natural hosts, but not on natural hosts. The molecular mechanisms that lead to the development of tick resistance are not fully understood, and both host and tick factors are invoked in this phenomenon. Advances in molecular tools to address the host and the tick are beginning to reveal new insights into this phenomenon and to uncover a deeper understanding of the fundamental biology of tick-host interactions. This review will focus on the expanding understanding of acquired tick resistance and highlight the impact of this understanding on anti-tick vaccine development efforts.


Asunto(s)
Proteoma/fisiología , Infestaciones por Garrapatas/inmunología , Garrapatas/fisiología , Animales , Modelos Animales de Enfermedad , Resistencia a la Enfermedad , Interacciones Huésped-Parásitos/inmunología , Humanos
4.
Infect Immun ; 88(12)2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-32928964

RESUMEN

Borrelia burgdorferi causes Lyme disease, the most common tick-transmitted illness in North America. When Ixodes scapularis feed on an infected vertebrate host, spirochetes enter the tick gut along with the bloodmeal and colonize the vector. Here, we show that a secreted tick protein, I. scapularisprotein disulfide isomerase A3 (IsPDIA3), enhances B. burgdorferi colonization of the tick gut. I. scapularis ticks in which ispdiA3 has been knocked down using RNA interference have decreased spirochete colonization of the tick gut after engorging on B. burgdorferi-infected mice. Moreover, administration of IsPDIA3 antiserum to B. burgdorferi-infected mice reduced the ability of spirochetes to colonize the tick when feeding on these animals. We show that IsPDIA3 modulates inflammatory responses at the tick bite site, potentially facilitating spirochete survival at the vector-host interface as it exits the vertebrate host to enter the tick gut. These data provide functional insights into the complex interactions between B. burgdorferi and its arthropod vector and suggest additional targets to interfere with the spirochete life cycle.


Asunto(s)
Borrelia burgdorferi/fisiología , Ixodes/metabolismo , Enfermedad de Lyme/transmisión , Proteína Disulfuro Isomerasas/metabolismo , Secuencia de Aminoácidos , Animales , Vectores Arácnidos/microbiología , Línea Celular , Técnicas de Silenciamiento del Gen , Humanos , Inmunidad Humoral , Inflamación/enzimología , Inflamación/genética , Inflamación/metabolismo , Ixodes/enzimología , Ixodes/genética , Proteínas de la Membrana/metabolismo , Ratones , Filogenia , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/inmunología , Interferencia de ARN , Proteínas Recombinantes , Alineación de Secuencia , Spirochaetales/fisiología
5.
Clin Infect Dis ; 70(8): 1768-1773, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-31620776

RESUMEN

Lyme disease, caused by some Borrelia burgdorferi sensu lato, is the most common tick-borne illness in the Northern Hemisphere and the number of cases, and geographic spread, continue to grow. Previously identified B. burgdorferi proteins, lipid immunogens, and live mutants lead the design of canonical vaccines aimed at disrupting infection in the host. Discovery of the mechanism of action of the first vaccine catalyzed the development of new strategies to control Lyme disease that bypassed direct vaccination of the human host. Thus, novel prevention concepts center on proteins produced by B. burgdorferi during tick transit and on tick proteins that mediate feeding and pathogen transmission. A burgeoning area of research is tick immunity as it can unlock mechanistic pathways that could be targeted for disruption. Studies that shed light on the mammalian immune pathways engaged during tick-transmitted B. burgdorferi infection would further development of vaccination strategies against Lyme disease.


Asunto(s)
Borrelia burgdorferi , Ixodes , Enfermedad de Lyme , Garrapatas , Vacunas , Animales , Humanos , Enfermedad de Lyme/prevención & control , Vacunación
6.
Proc Natl Acad Sci U S A ; 114(5): E781-E790, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28096373

RESUMEN

Arthropods transmit diverse infectious agents; however, the ways microbes influence their vector to enhance colonization are poorly understood. Ixodes scapularis ticks harbor numerous human pathogens, including Anaplasma phagocytophilum, the agent of human granulocytic anaplasmosis. We now demonstrate that A. phagocytophilum modifies the I. scapularis microbiota to more efficiently infect the tick. A. phagocytophilum induces ticks to express Ixodes scapularis antifreeze glycoprotein (iafgp), which encodes a protein with several properties, including the ability to alter bacterial biofilm formation. IAFGP thereby perturbs the tick gut microbiota, which influences the integrity of the peritrophic matrix and gut barrier-critical obstacles for Anaplasma colonization. Mechanistically, IAFGP binds the terminal d-alanine residue of the pentapeptide chain of bacterial peptidoglycan, resulting in altered permeability and the capacity of bacteria to form biofilms. These data elucidate the molecular mechanisms by which a human pathogen appropriates an arthropod antibacterial protein to alter the gut microbiota and more effectively colonize the vector.


Asunto(s)
Anaplasma phagocytophilum/fisiología , Microbioma Gastrointestinal , Interacciones Huésped-Patógeno , Ixodes/microbiología , Animales , Proteínas Anticongelantes/metabolismo , Proteínas de Artrópodos/metabolismo , Ehrlichiosis , Ratones , Peptidoglicano/metabolismo
7.
J Immunol ; 196(10): 4185-95, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27076681

RESUMEN

Borrelia miyamotoi is a relapsing fever spirochete in Ixodes ticks that has been recently identified as a human pathogen causing hard tick-borne relapsing fever (HTBRF) across the Northern Hemisphere. No validated serologic test exists, and current serologic assays have low sensitivity in early HTBRF. To examine the humoral immune response against B. miyamotoi, we infected C3H/HeN mice with B. miyamotoi strain LB-2001 expressing variable small protein 1 (Vsp1) and demonstrated that spirochetemia was cleared after 3 d, coinciding with anti-Vsp1 IgM production. Clearance was also observed after passive transfer of immune sera to infected SCID mice. Next, we showed that anti-Vsp1 IgG eliminates Vsp1-expressing B. miyamotoi, selecting for spirochetes expressing a variable large protein (VlpC2) resistant to anti-Vsp1. The viability of Asian isolate B. miyamotoi HT31, expressing Vlp15/16 and Vlp18, was also unaffected by anti-Vsp1. Finally, in nine HTBRF patients, we demonstrated IgM reactivity to Vsp1 in two and against Vlp15/16 in four ∼1 wk after these patients tested positive for B. miyamotoi by PCR. Our data show that B. miyamotoi is able to express various variable major proteins (VMPs) to evade humoral immunity and that VMPs are antigenic in humans. We propose that serologic tests based on VMPs are of additional value in diagnosing HTBRF.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Formación de Anticuerpos , Proteínas de la Membrana Bacteriana Externa/inmunología , Lipoproteínas/inmunología , Fiebre Recurrente/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Antibacterianos/sangre , Secuencia de Bases , Borrelia/inmunología , Femenino , Humanos , Inmunización Pasiva , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Ratones , Ratones Endogámicos C3H , Ratones SCID , Estructura Terciaria de Proteína
8.
PLoS Pathog ; 10(8): e1004278, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25102051

RESUMEN

Borrelia burgdorferi transmission to the vertebrate host commences with growth of the spirochete in the tick gut and migration from the gut to the salivary glands. This complex process, involving intimate interactions of the spirochete with the gut epithelium, is pivotal to transmission. We utilized a yeast surface display library of tick gut proteins to perform a global screen for tick gut proteins that might interact with Borrelia membrane proteins. A putative fibronectin type III domain-containing tick gut protein (Ixofin3D) was most frequently identified from this screen and prioritized for further analysis. Immunization against Ixofin3D and RNA interference-mediated reduction in expression of Ixofin3D resulted in decreased spirochete burden in tick salivary glands and in the murine host. Microscopic examination showed decreased aggregation of spirochetes on the gut epithelium concomitant with reduced expression of Ixofin3D. Our observations suggest that the interaction between Borrelia and Ixofin3D facilitates spirochete congregation to the gut during transmission, and provides a "molecular exit" direction for spirochete egress from the gut.


Asunto(s)
Proteínas de Artrópodos/metabolismo , Fibronectinas/metabolismo , Interacciones Huésped-Parásitos/fisiología , Enfermedad de Lyme/transmisión , Garrapatas/metabolismo , Animales , Borrelia burgdorferi , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Ratones , Microscopía Confocal , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa
9.
Emerg Infect Dis ; 20(7): 1183-90, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24960072

RESUMEN

Borrelia miyamotoi sensu lato, a relapsing fever Borrelia sp., is transmitted by the same ticks that transmit B. burgdorferi (the Lyme disease pathogen) and occurs in all Lyme disease-endemic areas of the United States. To determine the seroprevalence of IgG against B. miyamotoi sensu lato in the northeastern United States and assess whether serum from B. miyamotoi sensu lato-infected persons is reactive to B. burgdorferi antigens, we tested archived serum samples from area residents during 1991-2012. Of 639 samples from healthy persons, 25 were positive for B. miyamotoi sensu lato and 60 for B. burgdorferi. Samples from ≈10% of B. miyamotoi sensu lato-seropositive persons without a recent history of Lyme disease were seropositive for B. burgdorferi. Our results suggest that human B. miyamotoi sensu lato infection may be common in southern New England and that B. burgdorferi antibody testing is not an effective surrogate for detecting B. miyamotoi sensu lato infection.


Asunto(s)
Infecciones por Borrelia/epidemiología , Borrelia/inmunología , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Infecciones por Borrelia/sangre , Infecciones por Borrelia/inmunología , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Enfermedad de Lyme/sangre , Enfermedad de Lyme/epidemiología , Enfermedad de Lyme/inmunología , Masculino , Persona de Mediana Edad , New England/epidemiología , Estudios Seroepidemiológicos
10.
J Autoimmun ; 53: 85-94, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24767831

RESUMEN

The composition of the gut microbiome represents a very important environmental factor that influences the development of type 1 diabetes (T1D). We have previously shown that MyD88-deficient non-obese diabetic (MyD88-/-NOD) mice, that were protected from T1D development, had a different composition of gut microbiota compared to wild type NOD mice. The aim of our study was to investigate whether this protection could be transferred. We demonstrate that transfer of gut microbiota from diabetes-protected MyD88-deficient NOD mice, reduced insulitis and significantly delayed the onset of diabetes. Gut bacteria from MyD88-deficient mice, administered over a 3-week period, starting at 4 weeks of age, stably altered the family composition of the gut microbiome, with principally Lachnospiraceae and Clostridiaceae increased and Lactobacillaceae decreased. The transferred mice had a higher concentration of IgA and TGFß in the lumen that was accompanied by an increase in CD8(+)CD103(+) and CD8αß T cells in the lamina propria of the large intestine. These data indicate not only that gut bacterial composition can be altered after the neonatal/weaning period, but that the composition of the microbiome affects the mucosal immune system and can delay the development of autoimmune diabetes. This result has important implications for the development of probiotic treatment for T1D.


Asunto(s)
Bacterias/inmunología , Diabetes Mellitus Experimental , Intestinos , Microbiota/inmunología , Probióticos/farmacología , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/microbiología , Diabetes Mellitus Experimental/terapia , Intestinos/inmunología , Intestinos/microbiología , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología
11.
PLoS Pathog ; 7(6): e1002079, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21695244

RESUMEN

Borrelia burgdorferi, the causative agent of Lyme disease, is transmitted to humans by bite of Ixodes scapularis ticks. The mechanisms by which the bacterium is transmitted from vector to host are poorly understood. In this study, we show that the F(ab)(2) fragments of BBE31, a B.burgdorferi outer-surface lipoprotein, interfere with the migration of the spirochete from tick gut into the hemolymph during tick feeding. The decreased hemolymph infection results in lower salivary glands infection, and consequently attenuates mouse infection by tick-transmitted B. burgdorferi. Using a yeast surface display approach, a tick gut protein named TRE31 was identified to interact with BBE31. Silencing tre31 also decreased the B. burgdorferi burden in the tick hemolymph. Delineating the specific spirochete and arthropod ligands required for B. burgdorferi movement in the tick may lead to new strategies to interrupt the life cycle of the Lyme disease agent.


Asunto(s)
Borrelia burgdorferi/patogenicidad , Tracto Gastrointestinal/microbiología , Hemolinfa/microbiología , Enfermedad de Lyme/microbiología , Garrapatas/microbiología , Animales , Proteínas de la Membrana Bacteriana Externa , Lipoproteínas , Datos de Secuencia Molecular , Movimiento
12.
EMBO Rep ; 12(11): 1196-203, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21921936

RESUMEN

Ixodes ticks harbour several human pathogens belonging to the order Rickettsiales, including Anaplasma phagocytophilum, the agent of human anaplasmosis. When ticks feed on A. phagocytophilum-infected mice, the pathogen enters the ticks' gut. The bacteria then migrate from the gut to infect the salivary glands of the ticks and are transmitted to the next host via the saliva. The molecular mechanisms that enable the migration of A. phagocytophilum from the gut to the salivary glands are poorly understood. Here we show that a secreted tick protein, P11, is important in this process. We show that P11 enables A. phagocytophilum to infect tick haemocytes, which are required for the migration of A. phagocytophilum from the gut to the salivary glands. Silencing of p11 impaired the A. phagocytophilum infection of tick haemocytes in vivo and consequently decreased pathogen infection of the salivary glands. In vitro experiments showed that P11 could bind to A. phagocytophilum and thus facilitate its infection of tick cells. This report provides new insights into A. phagocytophilum infection of ticks and reveals new avenues to interrupt the life cycle of Anaplasma and related Rickettsial pathogens.


Asunto(s)
Anaplasma phagocytophilum/fisiología , Tracto Gastrointestinal/microbiología , Ixodes/inmunología , Proteínas Protozoarias/metabolismo , Glándulas Salivales/microbiología , Proteínas y Péptidos Salivales/metabolismo , Animales , Anticuerpos Antiprotozoarios/inmunología , Ehrlichiosis/microbiología , Conducta Alimentaria/fisiología , Regulación de la Expresión Génica , Silenciador del Gen , Hemocitos/microbiología , Hemolinfa/microbiología , Humanos , Ratones , Movimiento , Fagocitosis , Unión Proteica , Proteínas Protozoarias/inmunología , Proteínas y Péptidos Salivales/genética
13.
J Infect Dis ; 206(8): 1233-41, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22859824

RESUMEN

Ixodes scapularis transmits the agent of human granulocytic anaplasmosis, among other pathogens. The mechanisms used by the tick to control Anaplasma phagocytophilum are not known. We demonstrate that the I. scapularis Janus kinase (JAK)-signaling transducer activator of transcription (STAT) pathway plays a critical role in A. phagocytophilum infection of ticks. The A. phagocytophilum burden increases in salivary glands and hemolymph when the JAK-STAT pathway is suppressed by RNA interference. The JAK-STAT pathway exerts its anti-Anaplasma activity presumably through STAT-regulated effectors. A salivary gland gene family encoding 5.3-kDa antimicrobial peptides is highly induced upon A. phagocytophilum infection of tick salivary glands. Gene expression and electrophoretic mobility shift assays showed that the 5.3-kDa antimicrobial peptide-encoding genes are regulated by tick STAT. Silencing of these genes increased A. phagocytophilum infection of tick salivary glands and transmission to mammalian host. These data suggest that the JAK-STAT signaling pathway plays a key role in controlling A. phagocytophilum infection in ticks by regulating the expression of antimicrobial peptides.


Asunto(s)
Anaplasma phagocytophilum/inmunología , Péptidos Catiónicos Antimicrobianos/inmunología , Ixodes/microbiología , Janus Quinasa 1/inmunología , Factores de Transcripción STAT/inmunología , Transducción de Señal , Animales , Péptidos Catiónicos Antimicrobianos/biosíntesis , Ensayo de Cambio de Movilidad Electroforética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ixodes/inmunología , Glándulas Salivales/inmunología , Glándulas Salivales/microbiología
14.
Trends Parasitol ; 39(12): 1100-1113, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37838514

RESUMEN

Ticks are hematophagous arthropods that transmit disease-causing pathogens worldwide. Tick saliva deposited into the tick-bite site is composed of an array of immunomodulatory proteins that ensure successful feeding and pathogen transmission. These salivary proteins are often glycosylated, and glycosylation is potentially critical for the function of these proteins. Some salivary glycans are linked to the phenomenon of red meat allergy - an allergic response to red meat consumption in humans exposed to certain tick species. Tick salivary glycans are also invoked in the phenomenon of acquired tick resistance wherein non-natural host species exposed to tick bites develop an immune response that thwarts subsequent tick feeding. This review dwells on our current knowledge of these two phenomena, thematically linked by salivary glycans.


Asunto(s)
Hipersensibilidad a los Alimentos , Mordeduras de Garrapatas , Garrapatas , Humanos , Animales , Mordeduras de Garrapatas/complicaciones , Azúcares , Hipersensibilidad a los Alimentos/etiología , Polisacáridos
15.
Sci Transl Med ; 15(718): eadi7829, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37851823

RESUMEN

The deer tick transmits nearly half of the known tick-borne pathogens in the United States, and its expanding geographic range increases the risk of human infection. To decrease the abundance of and infection risk from deer ticks, approaches that include vaccines for human use and for animal hosts are desired.


Asunto(s)
Ixodes , Infestaciones por Garrapatas , Animales , Humanos
16.
Pathogens ; 12(1)2023 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-36678479

RESUMEN

The blacklegged tick, Ixodes scapularis, is the predominant vector of Borrelia burgdorferi, the agent of Lyme disease in the USA. Natural hosts of I. scapularis such as Peromyscus leucopus are repeatedly infested by these ticks without acquiring tick resistance. However, upon repeated tick infestations, non-natural hosts such as guinea pigs, mount a robust immune response against critical tick salivary antigens and acquire tick resistance able to thwart tick feeding and Borrelia burgdorferi transmission. The salivary targets of acquired tick resistance could serve as vaccine targets to prevent tick feeding and the tick transmission of human pathogens. Currently, there is no animal model able to demonstrate both tick resistance and diverse clinical manifestations of Lyme disease. Non-human primates serve as robust models of human Lyme disease. By evaluating the responses to repeated tick infestation, this animal model could accelerate our ability to define the tick salivary targets of acquired resistance that may serve as vaccines to prevent the tick transmission of human pathogens. Towards this goal, we assessed the development of acquired tick resistance in non-human primates upon repeated tick infestations. We report that following repeated tick infestations, non-human primates do not develop the hallmarks of acquired tick resistance observed in guinea pigs. However, repeated tick infestations elicit immune responses able to impair the tick transmission of B. burgdorferi. A mechanistic understanding of the protective immune responses will provide insights into B. burgdorferi-tick-host interactions and additionally contribute to anti-tick vaccine discovery.

17.
Vaccine ; 41(34): 4996-5002, 2023 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-37407406

RESUMEN

Acquired resistance to ticks can develop when animals are repeatedly exposed to ticks. Recently, acquired resistance to Ixodes scapularis was induced in guinea pigs immunized with an mRNA-lipid nanoparticle vaccine (19ISP) encoding 19 I. scapularis proteins. Here, we evaluated specific mRNAs present in 19ISP to identify critical components associated with resistance to ticks. A lipid nanoparticle containing 12 mRNAs which included all the targets within 19ISP that elicited strong humoral responses in guinea pigs, was sufficient to induce robust resistance to ticks. Lipid nanoparticles containing fewer mRNAs or a single mRNA were not able to generate strong resistance to ticks. All lipid nanoparticles containing salp14 mRNA, however, were associated with increased redness at the tick bite site - which is the first manifestation of acquired resistance to ticks. This study demonstrates that more than one I. scapularis target within 19ISP is required for resistance to ticks, and that additional targets may also play a role in this process.


Asunto(s)
Ixodes , Enfermedad de Lyme , Animales , Cobayas , ARN Mensajero , Ixodes/genética
18.
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
19.
J Exp Med ; 203(6): 1507-17, 2006 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-16717118

RESUMEN

Anaplasma phagocytophilum is the agent of human anaplasmosis, the second most common tick-borne illness in the United States. This pathogen, which is closely related to obligate intracellular organisms in the genera Rickettsia, Ehrlichia, and Anaplasma, persists in ticks and mammalian hosts; however, the mechanisms for survival in the arthropod are not known. We now show that A. phagocytophilum induces expression of the Ixodes scapularis salp16 gene in the arthropod salivary glands during vector engorgement. RNA interference-mediated silencing of salp16 gene expression interfered with the survival of A. phagocytophilum that entered ticks fed on A. phagocytophilum-infected mice. A. phagocytophilum migrated normally from A. phagocytophilum-infected mice to the gut of engorging salp16-deficient ticks, but up to 90% of the bacteria that entered the ticks were not able to successfully infect I. scapularis salivary glands. These data demonstrate the specific requirement of a pathogen for a tick salivary protein to persist within the arthropod and provide a paradigm for understanding how Rickettsia-like pathogens are maintained within vectors.


Asunto(s)
Anaplasma phagocytophilum/fisiología , Proteínas de Insectos/fisiología , Ixodes/microbiología , Glándulas Salivales/microbiología , Anaplasma phagocytophilum/genética , Animales , Secuencia de Bases , Ehrlichia/genética , Ehrlichia/fisiología , Mamíferos , Ratones , Ratones Endogámicos C3H , Interferencia de ARN , Rickettsia/genética , Rickettsia/fisiología , Infestaciones por Garrapatas/fisiopatología
20.
PLoS Pathog ; 6(11): e1001205, 2010 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21124826

RESUMEN

Ticks are distributed worldwide and affect human and animal health by transmitting diverse infectious agents. Effective vaccines against most tick-borne pathogens are not currently available. In this study, we characterized a tick histamine release factor (tHRF) from Ixodes scapularis and addressed the vaccine potential of this antigen in the context of tick engorgement and B. burgdorferi transmission. Results from western blotting and quantitative Reverse Transcription-PCR showed that tHRF is secreted in tick saliva, and upregulated in Borrelia burgdorferi-infected ticks. Further, the expression of tHRF was coincident with the rapid feeding phase of the tick, suggesting a role for tHRF in tick engorgement and concomitantly, for efficient B. burgdorferi transmission. Silencing tHRF by RNA interference (RNAi) significantly impaired tick feeding and decreased B. burgdorferi burden in mice. Interfering with tHRF by actively immunizing mice with recombinant tHRF, or passively transferring tHRF antiserum, also markedly reduced the efficiency of tick feeding and B. burgdorferi burden in mice. Recombinant tHRF was able to bind to host basophils and stimulate histamine release. Therefore, we speculate that tHRF might function in vivo to modulate vascular permeability and increase blood flow to the tick bite-site, facilitating tick engorgement. These findings suggest that blocking tHRF might offer a viable strategy to complement ongoing efforts to develop vaccines to block tick feeding and transmission of tick-borne pathogens.


Asunto(s)
Biomarcadores de Tumor/inmunología , Biomarcadores de Tumor/metabolismo , Borrelia burgdorferi/patogenicidad , Ixodes/fisiología , Enfermedad de Lyme/transmisión , Infestaciones por Garrapatas/microbiología , Animales , Biomarcadores de Tumor/antagonistas & inhibidores , Western Blotting , Conducta Alimentaria , Femenino , Histamina/metabolismo , Humanos , Inmunización , Enfermedad de Lyme/genética , Enfermedad de Lyme/metabolismo , Ratones , Ratones Endogámicos C3H , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Saliva/microbiología , Infestaciones por Garrapatas/inmunología , Proteína Tumoral Controlada Traslacionalmente 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA