Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Immunity ; 57(7): 1514-1532.e15, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38788712

RESUMEN

Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) functions as a critical stress sentinel that coordinates cell survival, inflammation, and immunogenic cell death (ICD). Although the catalytic function of RIPK1 is required to trigger cell death, its non-catalytic scaffold function mediates strong pro-survival signaling. Accordingly, cancer cells can hijack RIPK1 to block necroptosis and evade immune detection. We generated a small-molecule proteolysis-targeting chimera (PROTAC) that selectively degraded human and murine RIPK1. PROTAC-mediated depletion of RIPK1 deregulated TNFR1 and TLR3/4 signaling hubs, accentuating the output of NF-κB, MAPK, and IFN signaling. Additionally, RIPK1 degradation simultaneously promoted RIPK3 activation and necroptosis induction. We further demonstrated that RIPK1 degradation enhanced the immunostimulatory effects of radio- and immunotherapy by sensitizing cancer cells to treatment-induced TNF and interferons. This promoted ICD, antitumor immunity, and durable treatment responses. Consequently, targeting RIPK1 by PROTACs emerges as a promising approach to overcome radio- or immunotherapy resistance and enhance anticancer therapies.


Asunto(s)
Muerte Celular Inmunogénica , Proteolisis , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Transducción de Señal , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Humanos , Animales , Ratones , Proteolisis/efectos de los fármacos , Línea Celular Tumoral , Transducción de Señal/efectos de los fármacos , Muerte Celular Inmunogénica/efectos de los fármacos , Necroptosis/efectos de los fármacos , Necroptosis/inmunología , Neoplasias/inmunología , Neoplasias/tratamiento farmacológico , Ratones Endogámicos C57BL , Antineoplásicos/farmacología , Inmunoterapia/métodos
2.
Immunity ; 55(12): 2436-2453.e5, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36462503

RESUMEN

The factors that influence survival during severe infection are unclear. Extracellular chromatin drives pathology, but the mechanisms enabling its accumulation remain elusive. Here, we show that in murine sepsis models, splenocyte death interferes with chromatin clearance through the release of the DNase I inhibitor actin. Actin-mediated inhibition was compensated by upregulation of DNase I or the actin scavenger gelsolin. Splenocyte death and neutrophil extracellular trap (NET) clearance deficiencies were prevalent in individuals with severe COVID-19 pneumonia or microbial sepsis. Activity tracing by plasma proteomic profiling uncovered an association between low NET clearance and increased COVID-19 pathology and mortality. Low NET clearance activity with comparable proteome associations was prevalent in healthy donors with low-grade inflammation, implicating defective chromatin clearance in the development of cardiovascular disease and linking COVID-19 susceptibility to pre-existing conditions. Hence, the combination of aberrant chromatin release with defects in protective clearance mechanisms lead to poor survival outcomes.


Asunto(s)
COVID-19 , Sepsis , Animales , Ratones , Actinas , Cromatina , Desoxirribonucleasa I , ADN , Neutrófilos , Proteómica
3.
Proc Natl Acad Sci U S A ; 119(2)2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34996874

RESUMEN

Lethal toxin (LeTx)-mediated killing of myeloid cells is essential for Bacillus anthracis, the causative agent of anthrax, to establish systemic infection and induce lethal anthrax. The "LeTx-sensitive" NLRP1b inflammasome of BALB/c and 129S macrophages swiftly responds to LeTx intoxication with pyroptosis and secretion of interleukin (IL)-1ß. However, human NLRP1 is nonresponsive to LeTx, prompting us to investigate B. anthracis host-pathogen interactions in C57BL/6J (B6) macrophages and mice that also lack a LeTx-sensitive Nlrp1b allele. Unexpectedly, we found that LeTx intoxication and live B. anthracis infection of B6 macrophages elicited robust secretion of IL-1ß, which critically relied on the NLRP3 inflammasome. TNF signaling through both TNF receptor 1 (TNF-R1) and TNF-R2 were required for B. anthracis-induced NLRP3 inflammasome activation, which was further controlled by RIPK1 kinase activity and LeTx-mediated proteolytic inactivation of MAP kinase signaling. In addition to activating the NLRP3 inflammasome, LeTx-induced MAPKK inactivation and TNF production sensitized B. anthracis-infected macrophages to robust RIPK1- and caspase-8-dependent apoptosis. In agreement, purified LeTx triggered RIPK1 kinase activity- and caspase-8-dependent apoptosis only in macrophages primed with TNF or following engagement of TRIF-dependent Toll-like receptors. Consistently, genetic and pharmacological inhibition of RIPK1 inhibited NLRP3 inflammasome activation and apoptosis of LeTx-intoxicated and B. anthracis-infected macrophages. Caspase-8/RIPK3-deficient mice were significantly protected from B. anthracis-induced lethality, demonstrating the in vivo pathophysiological relevance of this cytotoxic mechanism. Collectively, these results establish TNF- and RIPK1 kinase activity-dependent NLRP3 inflammasome activation and macrophage apoptosis as key host-pathogen mechanisms in lethal anthrax.


Asunto(s)
Apoptosis , Bacillus anthracis/metabolismo , Caspasa 8/metabolismo , Inflamasomas/metabolismo , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Carbunco , Caspasa 8/genética , Interacciones Huésped-Patógeno/fisiología , Inflamasomas/genética , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Piroptosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Transducción de Señal
4.
Proc Natl Acad Sci U S A ; 113(50): 14384-14389, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27911804

RESUMEN

Familial Mediterranean fever (FMF) is the most common monogenic autoinflammatory disease worldwide. It is caused by mutations in the inflammasome adaptor Pyrin, but how FMF mutations alter signaling in FMF patients is unknown. Herein, we establish Clostridium difficile and its enterotoxin A (TcdA) as Pyrin-activating agents and show that wild-type and FMF Pyrin are differentially controlled by microtubules. Diverse microtubule assembly inhibitors prevented Pyrin-mediated caspase-1 activation and secretion of IL-1ß and IL-18 from mouse macrophages and human peripheral blood mononuclear cells (PBMCs). Remarkably, Pyrin inflammasome activation persisted upon microtubule disassembly in PBMCs of FMF patients but not in cells of patients afflicted with other autoinflammatory diseases. We further demonstrate that microtubules control Pyrin activation downstream of Pyrin dephosphorylation and that FMF mutations enable microtubule-independent assembly of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) micrometer-sized perinuclear structures (specks). The discovery that Pyrin mutations remove the obligatory requirement for microtubules in inflammasome activation provides a conceptual framework for understanding FMF and enables immunological screening of FMF mutations.


Asunto(s)
Fiebre Mediterránea Familiar/genética , Fiebre Mediterránea Familiar/metabolismo , Inflamasomas/metabolismo , Mutación , Pirina/genética , Pirina/metabolismo , Animales , Toxinas Bacterianas/toxicidad , Proteínas Adaptadoras de Señalización CARD/metabolismo , Infecciones por Clostridium/inmunología , Infecciones por Clostridium/metabolismo , Enterotoxinas/toxicidad , Fiebre Mediterránea Familiar/inmunología , Células HEK293 , Humanos , Inflamasomas/efectos de los fármacos , Inflamasomas/inmunología , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microtúbulos/efectos de los fármacos , Microtúbulos/inmunología , Microtúbulos/metabolismo , Pirina/inmunología , Tubulina (Proteína)/metabolismo
5.
Cell Mol Life Sci ; 73(11-12): 2335-47, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27048821

RESUMEN

Inflammasomes are multi-protein platforms that are organized in the cytosol to cope with pathogens and cellular stress. The pattern recognition receptors NLRP1, NLRP3, NLRC4, AIM2 and Pyrin all assemble canonical platforms for caspase-1 activation, while caspase-11-dependent inflammasomes respond to intracellular Gram-negative pathogens. Inflammasomes are chiefly known for their roles in maturation and secretion of the inflammatory cytokines interleukin-(IL)1ß and IL18, but they can also induce regulated cell death. Activation of caspases 1 and 11 in myeloid cells can trigger pyroptosis, a lytic and inflammatory cell death mode. Pyroptosis has been implicated in secretion of IL1ß, IL18 and intracellular alarmins. Akin to these factors, it may have beneficial roles in controlling pathogen replication, but become detrimental in the context of chronic autoinflammatory diseases. Inflammasomes are increasingly implicated in induction of additional regulated cell death modes such as pyronecrosis and apoptosis. In this review, we overview recent advances in inflammasome-associated cell death research, illustrating the polyvalent roles of these macromolecular platforms in regulated cell death signaling.


Asunto(s)
Apoptosis/fisiología , Caspasa 1/metabolismo , Bacterias Gramnegativas/inmunología , Inflamasomas/metabolismo , Piroptosis/fisiología , Alarminas/metabolismo , Animales , Caspasas/metabolismo , Caspasas Iniciadoras , Humanos , Inflamación/patología , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Noqueados , Transducción de Señal
6.
Elife ; 122024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39009040

RESUMEN

Background: Prinflammatory extracellular chromatin from neutrophil extracellular traps (NETs) and other cellular sources is found in COVID-19 patients and may promote pathology. We determined whether pulmonary administration of the endonuclease dornase alfa reduced systemic inflammation by clearing extracellular chromatin. Methods: Eligible patients were randomized (3:1) to the best available care including dexamethasone (R-BAC) or to BAC with twice-daily nebulized dornase alfa (R-BAC + DA) for seven days or until discharge. A 2:1 ratio of matched contemporary controls (CC-BAC) provided additional comparators. The primary endpoint was the improvement in C-reactive protein (CRP) over time, analyzed using a repeated-measures mixed model, adjusted for baseline factors. Results: We recruited 39 evaluable participants: 30 randomized to dornase alfa (R-BAC +DA), 9 randomized to BAC (R-BAC), and included 60 CC-BAC participants. Dornase alfa was well tolerated and reduced CRP by 33% compared to the combined BAC groups (T-BAC). Least squares (LS) mean post-dexamethasone CRP fell from 101.9 mg/L to 23.23 mg/L in R-BAC +DA participants versus a 99.5 mg/L to 34.82 mg/L reduction in the T-BAC group at 7 days; p=0.01. The anti-inflammatory effect of dornase alfa was further confirmed with subgroup and sensitivity analyses on randomised participants only, mitigating potential biases associated with the use of CC-BAC participants. Dornase alfa increased live discharge rates by 63% (HR 1.63, 95% CI 1.01-2.61, p=0.03), increased lymphocyte counts (LS mean: 1.08 vs 0.87, p=0.02) and reduced circulating cf-DNA and the coagulopathy marker D-dimer (LS mean: 570.78 vs 1656.96 µg/mL, p=0.004). Conclusions: Dornase alfa reduces pathogenic inflammation in COVID-19 pneumonia, demonstrating the benefit of cost-effective therapies that target extracellular chromatin. Funding: LifeArc, Breathing Matters, The Francis Crick Institute (CRUK, Medical Research Council, Wellcome Trust). Clinical trial number: NCT04359654.


Asunto(s)
Antiinflamatorios , Tratamiento Farmacológico de COVID-19 , COVID-19 , Desoxirribonucleasa I , Humanos , Masculino , Femenino , Desoxirribonucleasa I/administración & dosificación , Desoxirribonucleasa I/uso terapéutico , Persona de Mediana Edad , Antiinflamatorios/administración & dosificación , Antiinflamatorios/uso terapéutico , Anciano , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Trampas Extracelulares/efectos de los fármacos , SARS-CoV-2 , Proteína C-Reactiva/análisis , Proteína C-Reactiva/metabolismo , Dexametasona/administración & dosificación , Dexametasona/uso terapéutico , Adulto , Nebulizadores y Vaporizadores , Resultado del Tratamiento , Administración por Inhalación
7.
Nat Commun ; 13(1): 4658, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35945238

RESUMEN

The mechanisms linking systemic infection to hyperinflammation and immune dysfunction in sepsis are poorly understood. Extracellular histones promote sepsis pathology, but their source and mechanism of action remain unclear. Here, we show that by controlling fungi and bacteria captured by splenic macrophages, neutrophil-derived myeloperoxidase attenuates sepsis by suppressing histone release. In systemic candidiasis, microbial capture via the phagocytic receptor SIGNR1 neutralizes myeloperoxidase by facilitating marginal zone infiltration and T cell death-dependent histone release. Histones and hyphae induce cytokines in adjacent CD169 macrophages including G-CSF that selectively depletes mature Ly6Ghigh neutrophils by shortening their lifespan in favour of immature Ly6Glow neutrophils with a defective oxidative burst. In sepsis patient plasma, these mediators shorten mature neutrophil lifespan and correlate with neutrophil mortality markers. Consequently, high G-CSF levels and neutrophil lifespan shortening activity are associated with sepsis patient mortality. Hence, by exploiting phagocytic receptors, pathogens degrade innate and adaptive immunity through the detrimental impact of downstream effectors on neutrophil lifespan.


Asunto(s)
Neutrófilos , Sepsis , Factor Estimulante de Colonias de Granulocitos/metabolismo , Histonas/metabolismo , Humanos , Longevidad , Macrófagos/metabolismo , Peroxidasa/metabolismo , Linfocitos T/metabolismo
8.
Artículo en Inglés | MEDLINE | ID: mdl-31570336

RESUMEN

Inflammasomes assemble in the cytosol of myeloid and epithelial cells on sensing of cellular stress and pathogen-associated molecular patterns and serve as scaffolds for recruitment and activation of inflammatory caspases. Inflammasomes play beneficial roles in host and immune responses against diverse pathogens but may also promote inflammatory tissue damage if uncontrolled. Gasdermin D (GSDMD) is a recently identified substrate of murine caspase-1 and caspase-11, and human caspases-1, -4, and -5 that mediates a regulated lytic cell death mode termed pyroptosis. Recent studies have identified pyroptosis as a critical inflammasome effector mechanism that controls inflammasome-dependent cytokine secretion and contributes to antimicrobial defense and inflammasome-mediated autoinflammatory diseases. Here, we review recent developments on inflammasome-associated effector functions with an emphasis on the emerging roles of gasdermin pores and pyroptosis.


Asunto(s)
Caspasas/metabolismo , Inflamasomas/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Animales , Apoptosis , Caspasa 1/metabolismo , Caspasas Iniciadoras/metabolismo , Humanos , Inflamación , Interleucina-1/metabolismo , Interleucina-1beta/metabolismo , Ratones , Piroptosis
9.
Cell Rep ; 32(4): 107959, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32726624

RESUMEN

Pyroptosis has emerged as a key mechanism by which inflammasomes promote host defense against microbial pathogens and sterile inflammation. Gasdermin D (GSDMD)-mediated cell lysis is a hallmark of pyroptosis, but our understanding of cell death signaling during pyroptosis is fragmented. Here, we show that independently of GSDMD-mediated plasma membrane permeabilization, inflammasome receptors engage caspase-1 and caspase-8, both of which redundantly promote activation of apoptotic executioner caspase-3 and caspase-7 in pyroptotic macrophages. Impaired GSDMD pore formation downstream of caspase-1 and caspase-8 activation suffices to unmask the apoptotic phenotype of pyroptotic macrophages. Combined inactivation of initiator caspase-1 and caspase-8, or executioner caspase-3 and caspase-7, is required to abolish inflammasome-induced DEVDase activity during pyroptosis and in apoptotic Gsdmd-/- cells. Collectively, these results unveil a robust apoptotic caspase network that is activated in parallel to GSDMD-mediated plasma membrane permeabilization and safeguards cell death induction in pyroptotic macrophages.


Asunto(s)
Caspasas/metabolismo , Macrófagos/metabolismo , Piroptosis/fisiología , Animales , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/metabolismo , Caspasa 1/metabolismo , Caspasa 1/fisiología , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Caspasa 8/metabolismo , Caspasa 8/fisiología , Muerte Celular , Membrana Celular/metabolismo , Femenino , Inflamasomas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Unión a Fosfato/metabolismo
10.
Cell Death Differ ; 26(1): 146-161, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29666477

RESUMEN

Pyroptosis is rapidly emerging as a mechanism of anti-microbial host defense, and of extracellular release of the inflammasome-dependent cytokines interleukin (IL)-1ß and IL-18, which contributes to autoinflammatory pathology. Caspases 1, 4, 5 and 11 trigger this regulated form of necrosis by cleaving the pyroptosis effector gasdermin D (GSDMD), causing its pore-forming amino-terminal domain to oligomerize and perforate the plasma membrane. However, the subcellular events that precede pyroptotic cell lysis are ill defined. In this study, we triggered primary macrophages to undergo pyroptosis from three inflammasome types and recorded their dynamics and morphology using high-resolution live-cell spinning disk confocal laser microscopy. Based on quantitative analysis of single-cell subcellular events, we propose a model of pyroptotic cell disintegration that is initiated by opening of GSDMD-dependent ion channels or pores that are more restrictive than recently proposed GSDMD pores, followed by osmotic cell swelling, commitment of mitochondria and other membrane-bound organelles prior to sudden rupture of the plasma membrane and full permeability to intracellular proteins. This study provides a dynamic framework for understanding cellular changes that occur during pyroptosis, and charts a chronological sequence of GSDMD-mediated subcellular events that define pyroptotic cell death at the single-cell level.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/citología , Proteínas de Unión a Fosfato/metabolismo , Piroptosis/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Membrana Celular/metabolismo , Inflamasomas/metabolismo , Lisosomas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Necroptosis , Necrosis/metabolismo , Fosfatidilserinas/metabolismo , Piroptosis/genética , Análisis de la Célula Individual
11.
Life Sci Alliance ; 2(1)2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30718379

RESUMEN

Activating germline mutations in the human inflammasome sensor NLRP1 causes palmoplantar dyskeratosis and susceptibility to Mendelian autoinflammatory diseases. Recent studies have shown that the cytosolic serine dipeptidyl peptidases DPP8 and DPP9 suppress inflammasome activation upstream of NLRP1 and CARD8 in human keratinocytes and peripheral blood mononuclear cells. Moreover, pharmacological inhibition of DPP8/DPP9 protease activity was shown to induce pyroptosis in murine C57BL/6 macrophages without eliciting other inflammasome hallmark responses. Here, we show that DPP8/DPP9 inhibition in macrophages that express a Bacillus anthracis lethal toxin (LeTx)-sensitive Nlrp1b allele triggered significantly accelerated pyroptosis concomitant with caspase-1 maturation, ASC speck assembly, and secretion of mature IL-1ß and IL-18. Genetic ablation of ASC prevented DPP8/DPP9 inhibition-induced caspase-1 maturation and partially hampered pyroptosis and inflammasome-dependent cytokine release, whereas deletion of caspase-1 or gasdermin D triggered apoptosis in the absence of IL-1ß and IL-18 secretion. In conclusion, blockade of DPP8/DPP9 protease activity triggers rapid pyroptosis and canonical inflammasome hallmarks in primary macrophages that express a LeTx-responsive Nlrp1b allele.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/antagonistas & inhibidores , Inflamasomas/metabolismo , Macrófagos/metabolismo , Alelos , Animales , Antígenos Bacterianos , Apoptosis/efectos de los fármacos , Toxinas Bacterianas , Ácidos Borónicos/administración & dosificación , Ácidos Borónicos/farmacología , Proteínas Adaptadoras de Señalización CARD/genética , Caspasa 1/metabolismo , Línea Celular , Dipéptidos/administración & dosificación , Dipéptidos/farmacología , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Piroptosis/efectos de los fármacos
12.
Cell Rep ; 21(12): 3427-3444, 2017 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-29262324

RESUMEN

The caspase activation and recruitment domain (CARD)-based inflammasome sensors NLRP1b and NLRC4 induce caspase-1-dependent pyroptosis independent of the inflammasome adaptor ASC. Here, we show that NLRP1b and NLRC4 trigger caspase-8-mediated apoptosis as an alternative cell death program in caspase-1-/- macrophages and intestinal epithelial organoids (IECs). The caspase-8 adaptor FADD was recruited to ASC specks, which served as cytosolic platforms for caspase-8 activation and NLRP1b/NLRC4-induced apoptosis. We further found that caspase-1 protease activity dominated over scaffolding functions in suppressing caspase-8 activation and induction of apoptosis of macrophages and IECs. Moreover, TLR-induced c-FLIP expression inhibited caspase-8-mediated apoptosis downstream of ASC speck assembly, but did not affect pyroptosis induction by NLRP1b and NLRC4. Moreover, unlike during pyroptosis, NLRP1b- and NLRC4-elicited apoptosis retained alarmins and the inflammasome-matured cytokines interleukin 1ß (IL-1ß) and IL-18 intracellularly. This work identifies critical mechanisms regulating apoptosis induction by the inflammasome sensors NLRP1b and NLRC4 and suggests converting pyroptosis into apoptosis as a paradigm for suppressing inflammation.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Proteínas de Unión al Calcio/metabolismo , Caspasa 1/metabolismo , Inflamasomas/metabolismo , Piroptosis , Animales , Caspasa 8/metabolismo , Enterocitos/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores Toll-Like/metabolismo
13.
Ticks Tick Borne Dis ; 6(3): 364-75, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25800099

RESUMEN

Birds are important in the maintenance and spread of ticks and tick-borne diseases. In this context we screened birds in the Atlantic forest north of the São Francisco River and Caatinga in northeast Brazil. In the Atlantic forest Amblyomma longirostre, Amblyomma nodosum, Amblyomma varium and Amblyomma auricularium were identified. A. longirostre was infected by "Candidatus Rickettsia amblyommii" and A. nodosum by a Rickettsia parkeri-like agent. In Caatinga, Amblyomma parvum and A. auricularium were identified. A. auricularium was infected by "Ca. R. amblyommii" and Rickettsia bellii. "Canditatus Rickettsia andenae" was also identified in A. parvum collected from birds in Caatinga. In addition, Rickettsia sp. genotype AL was identified in A. varium collected on the clothes of the field team in one area of Atlantic forest. Here we provide a series of new host records for several Neotropical Amblyomma species and document rickettsial infections of "Ca. R. amblyomii" and a R. parkeri-like agent in Paraíba State, and R. bellii and "Ca. R. andenae" in Bahia State. For the first time we provide information regarding the infection of A. varium by "Ca. R. amblyommii".


Asunto(s)
Enfermedades de las Aves/epidemiología , Ixodidae/microbiología , Infecciones por Rickettsia/epidemiología , Rickettsia/aislamiento & purificación , Infestaciones por Garrapatas/veterinaria , Animales , Enfermedades de las Aves/microbiología , Aves , Brasil/epidemiología , Ecosistema , Femenino , Especificidad del Huésped , Larva , Masculino , Ninfa , Prevalencia , Rickettsia/genética , Infecciones por Rickettsia/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA