Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Virus Res ; 341: 199322, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38228190

RESUMEN

The emergence of highly infectious pathogens with their potential for triggering global pandemics necessitate the development of effective treatment strategies, including broad-spectrum antiviral therapies to safeguard human health. This study investigates the antiviral activity of emetine, dehydroemetine (DHE), and congeneric compounds against SARS-CoV-2 and HCoV-OC43, and evaluates their impact on the host cell. Concurrently, we assess the potential cardiotoxicity of these ipecac alkaloids. Significantly, our data reveal that emetine and the (-)-R,S isomer of 2,3-dehydroemetine (designated in this paper as DHE4) reduce viral growth at nanomolar concentrations (i.e., IC50 ∼ 50-100 nM), paralleling those required for inhibition of protein synthesis, while calcium channel blocking activity occurs at elevated concentrations (i.e., IC50 ∼ 40-60 µM). Our findings suggest that the antiviral mechanisms primarily involve disruption of host cell protein synthesis and is demonstrably stereoisomer specific. The prospect of a therapeutic window in which emetine or DHE4 inhibit viral propagation without cardiotoxicity renders these alkaloids viable candidates in strategies worthy of clinical investigation.


Asunto(s)
Alcaloides , Emetina , Emetina/análogos & derivados , Humanos , Emetina/farmacología , Ipeca/farmacología , Cardiotoxicidad , Antivirales/toxicidad
2.
J Med Entomol ; 61(2): 442-453, 2024 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-38104248

RESUMEN

With the introduction of siliconized artificial membranes, various artificial feeding systems (AFS) for hard ticks (Ixodidae) have been developed over the last decades. Most AFS utilize similar core components but employ diverse approaches, materials, and experimental conditions. Published work describes different combinations of the core components without experimental optimizations for the artificial feeding of different tick species. Amblyomma americanum L., (Acari: Ixodidae) (lone star tick) is a known vector and reservoir for diverse tick-borne pathogens, such as Rickettsia amblyommatis and Ehrlichia chaffeensis. Ongoing environmental changes have supported the expansion of A. americanum into new habitats, contributing to increased tick-borne diseases in endemic areas. However, a significant knowledge gap exists in understanding the underlying mechanisms involved in A. americanum interactions with tick-borne pathogens. Here, we performed a systematic analysis and developed an optimized AFS for nymphal lone star ticks. Our results demonstrate that Goldbeater's membranes, rabbit hair, hair extract, and adult lone star ticks significantly improved the attachment rate of nymphal ticks, whereas tick frass and frass extract did not. With the optimized conditions, we achieved an attachment rate of 46 ±â€…3% and a success rate of 100% (i.e., one or more attached ticks) in each feeding experiment for nymphal lone star ticks. When fed on sheep blood spiked with R. amblyommatis, both nymphal and adult lone star ticks acquired and maintained R. amblyommatis, demonstrating the feasibility of studying A. americanum-pathogen interactions using AFS. Our study can serve as a roadmap to optimize and improve AFS for other medically relevant tick species.


Asunto(s)
Ixodidae , Rickettsia , Rickettsiaceae , Conejos , Animales , Ovinos , Ixodidae/microbiología , Amblyomma , Rickettsiales , Ninfa/microbiología
3.
Infect Immun ; 91(10): e0026023, 2023 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-37725063

RESUMEN

Staphylococcus aureus is a highly infective Gram-positive bacterial pathogen that causes a wide range of diseases in both healthy and immunocompromised individuals. It can evade host immune defenses by expressing numerous virulence factors and toxins. Coupled with the inability of the human host to develop protective immunity against S. aureus, the emergence of antibiotic-resistant strains complicates treatment options. The non-canonical Sts phosphatases negatively regulate signaling pathways in varied immune cell types. To determine the role of the Sts proteins in regulating host responses to a Gram-positive microorganism, we investigated the response of mice lacking Sts expression to S. aureus infection. Herein, we demonstrate that Sts -/- animals are significantly resistant to lethal intravenous doses of S. aureus strain USA300. Resistance is characterized by significantly enhanced survival and accelerated bacterial clearance in multiple peripheral organs. Infected Sts -/- animals do not display increased levels of cytokines TNFα, IFNγ, and IL-6 in the spleen, liver, and kidney during the early stages of the infection, suggesting that a heightened pro-inflammatory response does not underlie the resistance phenotype. In vivo ablation of mononuclear phagocytes compromises the Sts -/- enhanced CFU clearance phenotype. Additionally, Sts -/- bone marrow-derived macrophages demonstrate significantly enhanced restriction of intracellular S. aureus following ex vivo infection. These results reveal the Sts enzymes to be critical regulators of host immunity to a virulent Gram-positive pathogen and identify them as therapeutic targets for optimizing host anti-microbial responses.


Asunto(s)
Monoéster Fosfórico Hidrolasas , Infecciones Estafilocócicas , Staphylococcus aureus , Animales , Humanos , Ratones , Macrófagos/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Transducción de Señal , Infecciones Estafilocócicas/genética
4.
mBio ; 14(4): e0138823, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37489888

RESUMEN

Powassan virus (POWV) is an emerging tick-borne Flavivirus that causes lethal encephalitis and long-term neurologic damage. Currently, there are no POWV therapeutics, licensed vaccines, or reverse genetics systems for producing infectious POWVs from recombinant DNA. Using a circular polymerase extension reaction (CPER), we generated recombinant LI9 (recLI9) POWVs with attenuating NS1 protein mutations and a recLI9-split-eGFP reporter virus. NS1 proteins are highly conserved glycoproteins that regulate replication, spread, and neurovirulence. POWV NS1 contains three putative N-linked glycosylation sites that we modified individually in infectious recLI9 mutants (N85Q, N208Q, and N224Q). NS1 glycosylation site mutations reduced replication kinetics and were attenuated, with 1-2 log decreases in titer. Severely attenuated recLI9-N224Q exhibited a 2- to 3-day delay in focal cell-to-cell spread and reduced NS1 secretion but was lethal when intracranially inoculated into suckling mice. However, footpad inoculation of recLI9-N224Q resulted in the survival of 80% of mice and demonstrated that NS1-N224Q mutations reduce POWV neuroinvasion in vivo. To monitor NS1 trafficking, we CPER fused a split GFP11-tag to the NS1 C-terminus and generated an infectious reporter virus, recLI9-NS1-GFP11. Cells infected with recLI9-NS1-GFP11 revealed NS1 trafficking in live cells and the novel formation of large NS1-lined intracellular vesicles. An infectious recLI9-NS1-GFP11 reporter virus permits real-time analysis of NS1 functions in POWV replication, assembly, and secretion and provides a platform for evaluating antiviral compounds. Collectively, our robust POWV reverse genetics system permits analysis of viral spread and neurovirulence determinants in vitro and in vivo and enables the rational genetic design of live attenuated POWV vaccines. IMPORTANCE Our findings newly establish a mechanism for genetically modifying Powassan viruses (POWVs), systematically defining pathogenic determinants and rationally designing live attenuated POWV vaccines. This initial study demonstrates that mutating POWV NS1 glycosylation sites attenuates POWV spread and neurovirulence in vitro and in vivo. Our findings validate a robust circular polymerase extension reaction approach as a mechanism for developing, and evaluating, attenuated genetically modified POWVs. We further designed an infectious GFP-tagged reporter POWV that permits us to monitor secretory trafficking of POWV in live cells, which can be applied to screen potential POWV replication inhibitors. This robust system for modifying POWVs provides the ability to define attenuating POWV mutations and create genetically attenuated recPOWV vaccines.


Asunto(s)
Enfermedades Transmisibles , Virus de la Encefalitis Transmitidos por Garrapatas , Humanos , Glicosilación , Genética Inversa , Piel
5.
Ticks Tick Borne Dis ; 14(2): 102088, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36436461

RESUMEN

Since its discovery in the United States in 2017, the Asian longhorned tick (Haemaphysalis longicornis) has been detected in most eastern states between Rhode Island and Georgia. Long Island, east of New York City, a recognized high-risk area for tick-borne diseases, is geographically close to New Jersey and New York sites where H. longicornis was originally found. However, extensive tick surveys conducted in 2018 did not identify H. longicornis on Long Island. In stark contrast, our 2022 tick survey suggests that H. longicornis has rapidly invaded and expanded in multiple surveying sites on Long Island (12 out of 17 sites). Overall, the relative abundance of H. longicornis was similar to that of lone star ticks, Amblyomma americanum, a previously recognized tick species abundantly present on Long Island. Interestingly, our survey suggests that H. longicornis has expanded within the Appalachian forest ecological zone of Long Island's north shore compared to the Pine Barrens located on the south shore of Long Island. The rapid invasion and expansion of H. longicornis into an insular environment are different from the historical invasion and expansion of two native tick species, Ixodes scapularis (blacklegged tick or deer tick) and A. americanum, in Long Island. The implications of H. longicornis transmitting or introducing tick-borne pathogens of public health importance remain unknown.


Asunto(s)
Ixodidae , Enfermedades por Picaduras de Garrapatas , Garrapatas , Animales , Estados Unidos , Ciudad de Nueva York , Georgia , Amblyomma
6.
Virulence ; 13(1): 1752-1771, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36208040

RESUMEN

Rickettsiae include diverse Gram-negative microbial species that exhibit obligatory intracellular lifecycles between mammalian hosts and arthropod vectors. Human infections with arthropod-borne Rickettsia continue to cause significant morbidity and mortality as recent environmental changes foster the proliferation of arthropod vectors and increased exposure to humans. However, the technical difficulties in working with Rickettsia have delayed our progress in understanding the molecular mechanisms involved in rickettsial pathogenesis and disease transmission. Recent advances in developing genetic tools for Rickettsia have enabled investigators to identify virulence genes, uncover molecular functions, and characterize host responses to rickettsial determinants. Therefore, continued efforts to determine virulence genes and their biological functions will help us understand the underlying mechanisms associated with arthropod-borne rickettsioses.


Asunto(s)
Infecciones por Rickettsia , Rickettsia , Animales , Humanos , Mamíferos , Rickettsia/genética , Infecciones por Rickettsia/microbiología , Virulencia
7.
Infect Immun ; 90(9): e0062121, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35993770

RESUMEN

Ticks are hematophagous ectoparasites capable of transmitting multiple human pathogens. Environmental changes have supported the expansion of ticks into new geographical areas that have become the epicenters of tick-borne diseases (TBDs). The spotted fever group (SFG) of Rickettsia frequently infects ticks and causes tick-transmitted rickettsioses in areas of endemicity where ixodid ticks support host transmission during blood feeding. Ticks also serve as a reservoir for SFG Rickettsia. Among the members of SFG Rickettsia, R. rickettsii causes Rocky Mountain spotted fever (RMSF), the most lethal TBD in the United States. Cases of RMSF have been reported for over a century in association with several species of ticks in the United States. However, the isolation of R. rickettsii from ticks has decreased, and recent serological and epidemiological studies suggest that novel species of SFG Rickettsia are responsible for the increased number of cases of RMSF-like rickettsioses in the United States. Recent analyses of rickettsial genomes and advances in genetic and molecular studies of Rickettsia provided insights into the biology of Rickettsia with the identification of conserved and unique putative virulence genes involved in the rickettsial life cycle. Thus, understanding Rickettsia-host-tick interactions mediating successful disease transmission and pathogenesis for SFG rickettsiae remains an active area of research. This review summarizes recent advances in understanding how SFG Rickettsia species coopt and manipulate ticks and mammalian hosts to cause rickettsioses, with a particular emphasis on newly described or emerging SFG Rickettsia species.


Asunto(s)
Ixodidae , Infecciones por Rickettsia , Rickettsia , Fiebre Maculosa de las Montañas Rocosas , Garrapatas , Animales , Humanos , Ixodidae/microbiología , Mamíferos , Rickettsia/genética , Fiebre Maculosa de las Montañas Rocosas/microbiología , Garrapatas/microbiología
8.
J Virol ; 96(1): e0168221, 2022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-34643436

RESUMEN

Powassan viruses (POWVs) are neurovirulent tick-borne flaviviruses emerging in the northeastern United States, with a 2% prevalence in Long Island (LI) deer ticks (Ixodes scapularis). POWVs are transmitted within as little as 15 min of a tick bite and enter the central nervous system (CNS) to cause encephalitis (10% of cases are fatal) and long-term neuronal damage. POWV-LI9 and POWV-LI41 present in LI Ixodes ticks were isolated by directly inoculating VeroE6 cells with tick homogenates and detecting POWV-infected cells by immunoperoxidase staining. Inoculated POWV-LI9 and LI41 were exclusively present in infected cell foci, indicative of cell to cell spread, despite growth in liquid culture without an overlay. Cloning and sequencing establish POWV-LI9 as a phylogenetically distinct lineage II POWV strain circulating in LI deer ticks. Primary human brain microvascular endothelial cells (hBMECs) and pericytes form a neurovascular complex that restricts entry into the CNS. We found that POWV-LI9 and -LI41 and lineage I POWV-LB productively infect hBMECs and pericytes and that POWVs were basolaterally transmitted from hBMECs to lower-chamber pericytes without permeabilizing polarized hBMECs. Synchronous POWV-LI9 infection of hBMECs and pericytes induced proinflammatory chemokines, interferon-ß (IFN-ß) and proteins of the IFN-stimulated gene family (ISGs), with delayed IFN-ß secretion by infected pericytes. IFN inhibited POWV infection, but despite IFN secretion, a subset of POWV-infected hBMECs and pericytes remained persistently infected. These findings suggest a potential mechanism for POWVs (LI9/LI41 and LB) to infect hBMECs, spread basolaterally to pericytes, and enter the CNS. hBMEC and pericyte responses to POWV infection suggest a role for immunopathology in POWV neurovirulence and potential therapeutic targets for preventing POWV spread to neuronal compartments. IMPORTANCE We isolated POWVs from LI deer ticks (I. scapularis) directly in VeroE6 cells, and sequencing revealed POWV-LI9 as a distinct lineage II POWV strain. Remarkably, inoculation of VeroE6 cells with POWV-containing tick homogenates resulted in infected cell foci in liquid culture, consistent with cell-to-cell spread. POWV-LI9 and -LI41 and lineage I POWV-LB strains infected hBMECs and pericytes that comprise neurovascular complexes. POWVs were nonlytically transmitted basolaterally from infected hBMECs to lower-chamber pericytes, suggesting a mechanism for POWV transmission across the blood-brain barrier (BBB). POWV-LI9 elicited inflammatory responses from infected hBMEC and pericytes that may contribute to immune cell recruitment and neuropathogenesis. This study reveals a potential mechanism for POWVs to enter the CNS by infecting hBMECs and spreading basolaterally to abluminal pericytes. Our findings reveal that POWV-LI9 persists in cells that form a neurovascular complex spanning the BBB and suggest potential therapeutic targets for preventing POWV spread to neuronal compartments.


Asunto(s)
Vectores de Enfermedades , Virus de la Encefalitis Transmitidos por Garrapatas/fisiología , Encefalitis Transmitida por Garrapatas/virología , Ixodes/virología , Animales , Células Cultivadas , Virus de la Encefalitis Transmitidos por Garrapatas/clasificación , Virus de la Encefalitis Transmitidos por Garrapatas/efectos de los fármacos , Virus de la Encefalitis Transmitidos por Garrapatas/aislamiento & purificación , Encefalitis Transmitida por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/transmisión , Células Endoteliales , Orden Génico , Genoma Viral , Interacciones Huésped-Patógeno/inmunología , Humanos , Interferones/farmacología , Pericitos/virología , Filogenia , Replicación Viral/efectos de los fármacos
9.
mBio ; 12(6): e0275621, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34724828

RESUMEN

Outbreaks of emerging viral pathogens like severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are a major medical challenge. There is a pressing need for antivirals that can be rapidly deployed to curb infection and dissemination. We determined the efficacy of interferon lambda-1 (IFN-λ) as a broad-spectrum antiviral agent to inhibit SARS-CoV-2 infection and reduce pathology in a mouse model of disease. IFN-λ significantly limited SARS-CoV-2 production in primary human bronchial epithelial cells in culture. Pretreatment of human lung cells with IFN-λ completely blocked infectious virus production, and treatment with IFN-λ at the time of infection inhibited virus production more than 10-fold. To interrogate the protective effects of IFN-λ in response to SARS-CoV-2 infection, transgenic mice expressing the human angiotensin-converting enzyme 2 (ACE-2) were tested. One dose of IFN-λ administered intranasally was found to reduce animal morbidity and mortality. Our study with SARS-CoV-2 also revealed a sex differential in disease outcome. Male mice had higher mortality, reflecting the more severe symptoms and mortality found in male patients infected with SARS-CoV-2. The results indicate that IFN-λ potentially can treat early stages of SARS-CoV-2 infection and decrease pathology, and this murine model can be used to investigate the sex differential documented in COVID-19. IMPORTANCE The COVID-19 pandemic has claimed millions of lives worldwide. In this report, we used a preclinical mouse model to investigate the prophylactic and therapeutic value of intranasal IFN-λ for this acute respiratory disease. Specific vaccines have been responsible for curbing the transmission of SARS-CoV-2 in developed nations. However, vaccines require time to generate and keep pace with antigenic variants. There is a need for broad-spectrum prophylactic and therapeutic agents to combat new emerging viral pathogens. Our mouse model suggests IFN-λ has clinical utility, and it reflects the well-documented finding that male COVID-19 patients manifest more severe symptoms and mortality. Understanding this sex bias is critical for considering therapeutic approaches to COVID-19.


Asunto(s)
Antivirales/uso terapéutico , COVID-19/inmunología , COVID-19/terapia , Células Epiteliales/efectos de los fármacos , Interferones/inmunología , Interferones/farmacología , SARS-CoV-2/inmunología , Administración Intranasal , Enzima Convertidora de Angiotensina 2/genética , Animales , Antivirales/farmacología , Bronquios/citología , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Células Epiteliales/virología , Femenino , Células HEK293 , Humanos , Interferones/clasificación , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Transgénicos , Factores de Riesgo , SARS-CoV-2/efectos de los fármacos , Factores Sexuales
10.
Vaccine ; 39(29): 3907-3915, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34088508

RESUMEN

Exposure to Staphylococcus aureus does not lead to immunity as evidenced by the persistent colonization of one third of the human population. S. aureus immune escape is mediated by factors that preempt complement activation, destroy phagocytes, and modify B and T cell responses. One such factor, Staphylococcal protein A (SpA) encompasses five Immunoglobulin binding domains (IgBDs) that associate with the Fcγ domain to block phagocytosis. IgBDs also associate with Fab encoded by VH3 clan related genes. SpA binding to VH3-IgM that serves as a B cell receptor results in B cell expansion and secretion of antibodies with no specificity for S. aureus. SpA crosslinking of VH3-IgG and VH3-IgE bound to cognate receptors of mast cells and basophils promotes histamine release and anaphylaxis. Earlier work developed a prototype variant SpAKKAA with four amino acid substitutions in each IgBD. When tested in animal models, SpAKKAA elicited neutralizing antibodies and protection against infection. We show here that SpAKKAA retains crosslinking activity for VH3-IgG and VH3-IgE. We use a rational approach to design and test 67 new SpA variants for loss of VH3 binding and anaphylactic activities. We identify two detoxified candidates that elicit SpA-neutralizing antibodies and protect animals from S. aureus colonization and bloodstream infection. The new detoxified SpA candidates bear three instead of four amino acid substitutions thus increasing the development of SpA-specific antibodies. We propose that detoxified SpA variants unable to crosslink VH3-idiotypic immunoglobulin may be suitably developed as clinical-grade vaccines for safety and efficacy testing in humans.


Asunto(s)
Infecciones Estafilocócicas , Vacunas , Animales , Anticuerpos Neutralizantes , Humanos , Infecciones Estafilocócicas/prevención & control , Proteína Estafilocócica A/genética , Staphylococcus aureus/genética
11.
mBio ; 12(2)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33879590

RESUMEN

Staphylococcus aureus causes reiterative and chronic persistent infections. This can be explained by the formidable ability of this pathogen to escape immune surveillance mechanisms. Cells of S. aureus display the abundant staphylococcal protein A (SpA). SpA binds to immunoglobulin (Ig) molecules and coats the bacterial surface to prevent phagocytic uptake. SpA also binds and cross-links variable heavy 3 (VH3) idiotype (IgM) B cell receptors, promoting B cell expansion and the secretion of nonspecific VH3-IgM via a mechanism requiring CD4+ T cell help. SpA binding to antibodies is mediated by the N-terminal Ig-binding domains (IgBDs). The so-called region X, uncharacterized LysM domain, and C-terminal LPXTG sorting signal for peptidoglycan attachment complete the linear structure of the protein. Here, we report that both the LysM domain and the LPXTG motif sorting signal are required for the B cell superantigen activity of SpA in a mouse model of infection. SpA molecules purified from staphylococcal cultures are sufficient to exert B cell superantigen activity and promote immunoglobulin secretion as long as they carry intact LysM and LPXTG motif domains with bound peptidoglycan fragments. The LysM domain binds the glycan chains of peptidoglycan fragments, whereas the LPXTG motif is covalently linked to wall peptides lacking glycan. These findings emphasize the complexity of SpA interactions with B cell receptors.IMPORTANCE The LysM domain is found in all kingdoms of life. While their function in mammals is not known, LysM domains of bacteria and their phage parasites are associated with enzymes that cleave or remodel peptidoglycan. Plants recognize microbe-associated molecular patterns such as chitin via receptors endowed with LysM-containing ectodomains. In plants, such receptors play equally important roles in defense and symbiosis signaling. SpA of S. aureus carries a LysM domain that binds glycan strands of peptidoglycan to influence defined B cell responses that divert pathogen-specific adaptive immune responses.


Asunto(s)
Linfocitos B/inmunología , Peptidoglicano/inmunología , Peptidoglicano/metabolismo , Proteína Estafilocócica A/inmunología , Proteína Estafilocócica A/metabolismo , Staphylococcus aureus/inmunología , Superantígenos/inmunología , Inmunidad Adaptativa , Animales , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Polisacáridos/inmunología , Polisacáridos/metabolismo , Unión Proteica , Receptores de Antígenos de Linfocitos B , Proteína Estafilocócica A/genética
12.
Pathog Dis ; 79(5)2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33908603

RESUMEN

Rickettsia amblyommatis belongs to the spotted fever group of Rickettsia and infects Amblyomma americanum (Lone Star ticks) for transmission to offspring and mammals. Historically, the geographic range of A. americanum was restricted to the southeastern USA. However, recent tick surveys identified the progressive northward invasion of A. americanum, contributing to the increased number of patients with febrile illnesses of unknown etiology after a tick bite in the northeastern USA. While serological evidence strongly suggests that patients are infected with R. amblyommatis, the virulence potential of R. amblyommatis is not well established. Here, we performed a bioinformatic analysis of three genome sequences of R. amblyommatis and identified the presence of multiple putative virulence genes whose products are implicated for spotted fever pathogenesis. Similar to other pathogenic spotted fever rickettsiae, R. amblyommatis replicated intracellularly within the cytoplasm of tissue culture cells. Interestingly, R. amblyommatis displayed defective attachment to microvascular endothelial cells. The attachment defect and slow growth rate of R. amblyommatis required relatively high intravenous infectious doses to produce dose-dependent morbidity and mortality in C3H mice. In summary, our results corroborate clinical evidence that R. amblyommatis can cause mild disease manifestation in some patients.


Asunto(s)
Genoma Bacteriano/genética , Rickettsia/genética , Rickettsia/patogenicidad , Rickettsiosis Exantemáticas/microbiología , Animales , Línea Celular , Chlorocebus aethiops , Modelos Animales de Enfermedad , Células Endoteliales/microbiología , Genómica , Humanos , Ratones , Ratones Endogámicos C3H , Células Vero , Virulencia
13.
Proc Natl Acad Sci U S A ; 117(37): 22992-23000, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32855300

RESUMEN

Antibodies may bind to bacterial pathogens or their toxins to control infections, and their effector activity is mediated through the recruitment of complement component C1q or the engagement with Fcγ receptors (FcγRs). For bacterial pathogens that rely on a single toxin to cause disease, immunity correlates with toxin neutralization. Most other bacterial pathogens, including Staphylococcus aureus, secrete numerous toxins and evolved multiple mechanisms to escape opsonization and complement killing. Several vaccine candidates targeting defined surface antigens of S. aureus have failed to meet clinical endpoints. It is unclear that such failures can be solely attributed to the poor selection of antibody targets. Thus far, studies to delineate antibody-mediated uptake and killing of Gram-positive pathogens remain extremely limited. Here, we exploit 3F6-hIgG1, a human monoclonal antibody that binds and neutralizes the abundant surface-exposed Staphylococcal protein A (SpA). We find that galactosylation of 3F6-hIgG1 that favors C1q recruitment is indispensable for opsonophagocytic killing of staphylococci and for protection against bloodstream infection in animals. However, the simple removal of fucosyl residues, which results in reduced C1q binding and increased engagement with FcγR, maintains the opsonophagocytic killing and protective attributes of the antibody. We confirm these results by engineering 3F6-hIgG1 variants with biased binding toward C1q or FcγRs. While the therapeutic benefit of monoclonal antibodies against infectious disease agents may be debatable, the functional characterization of such antibodies represents a powerful tool for the development of correlates of protection that may guide future vaccine trials.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Fagocitosis/inmunología , Proteína Estafilocócica A/inmunología , Animales , Línea Celular , Glicosilación , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología
14.
Nature ; 574(7776): 57-62, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31534221

RESUMEN

The causative agent of plague, Yersinia pestis, uses a type III secretion system to selectively destroy immune cells in humans, thus enabling Y. pestis to reproduce in the bloodstream and be transmitted to new hosts through fleabites. The host factors that are responsible for the selective destruction of immune cells by plague bacteria are unknown. Here we show that LcrV, the needle cap protein of the Y. pestis type III secretion system, binds to the N-formylpeptide receptor (FPR1) on human immune cells to promote the translocation of bacterial effectors. Plague infection in mice is characterized by high mortality; however, Fpr1-deficient mice have increased survival and antibody responses that are protective against plague. We identified FPR1R190W as a candidate resistance allele in humans that protects neutrophils from destruction by the Y. pestis type III secretion system. Thus, FPR1 is a plague receptor on immune cells in both humans and mice, and its absence or mutation provides protection against Y. pestis. Furthermore, plague selection of FPR1 alleles appears to have shaped human immune responses towards other infectious diseases and malignant neoplasms.


Asunto(s)
Macrófagos/metabolismo , Neutrófilos/metabolismo , Peste/microbiología , Receptores de Formil Péptido/metabolismo , Yersinia pestis/metabolismo , Alelos , Animales , Antígenos Bacterianos/metabolismo , Adhesión Bacteriana , Sistemas CRISPR-Cas , Quimiotaxis/inmunología , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/citología , Neutrófilos/inmunología , Neutrófilos/microbiología , Peste/inmunología , Peste/prevención & control , Polimorfismo de Nucleótido Simple/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptores de Formil Péptido/antagonistas & inhibidores , Receptores de Formil Péptido/deficiencia , Receptores de Formil Péptido/genética , Sistemas de Secreción Tipo III/efectos de los fármacos , Células U937 , Yersinia pestis/química , Yersinia pestis/inmunología , Yersinia pestis/patogenicidad
15.
Proc Natl Acad Sci U S A ; 116(39): 19659-19664, 2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31413191

RESUMEN

Rickettsial diseases have long been diagnosed with serum antibodies cross-reactive against Proteus vulgaris (Weil-Felix reaction). Although Weil-Felix antibodies are associated with the development of immunity, their rickettsial target and contribution to disease pathogenesis are not established. Here, we developed a transposon for insertional mutagenesis of Rickettsia conorii, isolating variants defective for replication in cultured cells and in spotted fever pathogenesis. Mutations in the polysaccharide synthesis operon (pso) abolish lipopolysaccharide O-antigen synthesis and Weil-Felix serology and alter outer-membrane protein assembly. Unlike wild-type R. conorii, pso mutants cannot elicit bactericidal antibodies that bind O antigen. The pso operon is conserved among rickettsial pathogens, suggesting that bactericidal antibodies targeting O antigen may generate universal immunity that could be exploited to develop vaccines against rickettsial diseases.


Asunto(s)
Reacciones Cruzadas/inmunología , Antígenos O/inmunología , Rickettsia conorii/inmunología , Antibacterianos , Anticuerpos Antibacterianos/inmunología , Lipopolisacáridos/inmunología , Rickettsia/inmunología , Rickettsia/patogenicidad , Infecciones por Rickettsia/inmunología , Rickettsia conorii/patogenicidad
16.
Microbes Infect ; 19(2): 101-109, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28012900

RESUMEN

Humans and animals are colonized by members of the genus Staphylococcus, however only some of these species evolved to cause invasive disease. The genetic basis for conversion of commensal staphylococci into pathogens is not known. We hypothesized that Staphylococcus aureus genes for coagulation and agglutination in vertebrate blood (coa, vwb and clfA) may support pathogenic conversion. Expression of coa and vwb in Staphylococcus epidermidis or Staphylococcus simulans supported a coagulase-positive phenotype but not the ability to cause disease in a mouse model of bloodstream infection. However, the simultaneous expression of coa, vwb and clfA in coagulase-negative staphylococci enabled bacterial agglutination in plasma and enhanced survival of S. simulans in human whole blood. Agglutination of S. simulans in the bloodstream of infected mice upon expression of coa, vwb and clfA provided also a mean for dissemination and replication in distal organs. Thus, the acquisition of genes for bacterial agglutination with fibrin appear sufficient for the conversion of commensal staphylococci into invasive pathogens.


Asunto(s)
Bacteriemia/microbiología , Staphylococcus/patogenicidad , Factores de Virulencia/metabolismo , Animales , Coagulasa/genética , Coagulasa/metabolismo , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos BALB C , Staphylococcus/genética , Virulencia , Factores de Virulencia/genética
17.
Proc Natl Acad Sci U S A ; 113(20): 5718-23, 2016 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-27140614

RESUMEN

A hallmark of Staphylococcus aureus disease in humans is persistent infections without development of protective immune responses. Infected patients generate VH3 plasmablast expansions and increased VH3 idiotype Ig; however, the mechanisms for staphylococcal modification of immune responses are not known. We report here that S. aureus-infected mice generate VH3 antibody expansions via a mechanism requiring MHC-restricted antigen presentation to CD4(+) T cells and staphylococcal protein A (SpA), a cell wall-anchored surface molecule that binds Fcγ and VH3 variant heavy chains of Ig. VH3 expansion occurred with peptidoglycan-linked SpA from the bacterial envelope but not with recombinant SpA, and optimally required five tandem repeats of its Ig-binding domains. Signaling via receptor-interacting serine/threonine protein kinase 2 (RIPK2) was essential for implementing peptidoglycan-linked SpA superantigen activity. VH3 clan IgG from S. aureus-infected or SpA-treated animals was not pathogen-specific, suggesting that SpA cross-linking of VH3 idiotype B-cell receptors and activation via attached peptidoglycan are the determinants of staphylococcal escape from adaptive immune responses.


Asunto(s)
Anticuerpos Antibacterianos/biosíntesis , Infecciones Estafilocócicas/inmunología , Proteína Estafilocócica A/inmunología , Staphylococcus aureus/inmunología , Linfocitos T/metabolismo , Animales , Anticuerpos Antibacterianos/sangre , Formación de Anticuerpos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Peptidoglicano/inmunología , Infecciones Estafilocócicas/sangre , Infecciones Estafilocócicas/microbiología , Linfocitos T/inmunología , Linfocitos T/microbiología
18.
J Exp Med ; 213(3): 293-301, 2016 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-26880578

RESUMEN

Host immunity against bacteria typically involves antibodies that recognize the microbial surface and promote phagocytic killing. Methicillin-resistant Staphylococcus aureus (MRSA) is a frequent cause of lethal bloodstream infection; however, vaccines and antibody therapeutics targeting staphylococcal surface molecules have thus far failed to achieve clinical efficacy. S. aureus secretes coagulase (Coa), which activates host prothrombin and generates fibrin fibrils that protect the pathogen against phagocytosis by immune cells. Because of negative selection, the coding sequence for the prothrombin-binding D1-D2 domain is highly variable and does not elicit cross-protective immune responses. The R domain, tandem repeats of a 27-residue peptide that bind fibrinogen, is conserved at the C terminus of all Coa molecules, but its functional significance is not known. We show here that the R domain enables bloodstream infections by directing fibrinogen to the staphylococcal surface, generating a protective fibrin shield that inhibits phagocytosis. The fibrin shield can be marked with R-specific antibodies, which trigger phagocytic killing of staphylococci and protect mice against lethal bloodstream infections caused by a broad spectrum of MRSA isolates. These findings emphasize the critical role of coagulase in staphylococcal escape from opsonophagocytic killing and as a protective antigen for S. aureus vaccines.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Coagulasa/inmunología , Fagocitosis , Staphylococcus aureus/inmunología , Aglutinación , Animales , Anticuerpos Monoclonales/inmunología , Coagulasa/química , Femenino , Fibrina/metabolismo , Humanos , Ratones Endogámicos BALB C , Proteínas Opsoninas/metabolismo , Estructura Terciaria de Proteína , Secuencias Repetitivas de Aminoácido , Infecciones Estafilocócicas/sangre , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/prevención & control
19.
Nat Rev Microbiol ; 13(9): 529-43, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26272408

RESUMEN

Staphylococcus aureus, a bacterial commensal of the human nares and skin, is a frequent cause of soft tissue and bloodstream infections. A hallmark of staphylococcal infections is their frequent recurrence, even when treated with antibiotics and surgical intervention, which demonstrates the bacterium's ability to manipulate innate and adaptive immune responses. In this Review, we highlight how S. aureus virulence factors inhibit complement activation, block and destroy phagocytic cells and modify host B cell and T cell responses, and we discuss how these insights might be useful for the development of novel therapies against infections with antibiotic resistant strains such as methicillin-resistant S. aureus.


Asunto(s)
Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Aglutinación/inmunología , Activación de Complemento , Regulación Bacteriana de la Expresión Génica/fisiología , Humanos , Evasión Inmune , Activación de Linfocitos , Fagocitosis , Factores de Virulencia/metabolismo
20.
mBio ; 6(1)2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25564466

RESUMEN

UNLABELLED: Staphylococcus aureus infection is not associated with the development of protective immunity, and disease relapses occur frequently. We hypothesize that protein A, a factor that binds immunoglobulin Fcγ and cross-links V(H)3 clan B cell receptors (IgM), is the staphylococcal determinant for host immune suppression. To test this, vertebrate IgM was examined for protein A cross-linking. High V(H)3 binding activity occurred with human and guinea immunoglobulin, whereas mouse and rabbit immunoglobulins displayed little and no binding, respectively. Establishing a guinea pig model of S. aureus bloodstream infection, we show that protein A functions as a virulence determinant and suppresses host B cell responses. Immunization with SpA(KKAA), which cannot bind immunoglobulin, elicits neutralizing antibodies that enable guinea pigs to develop protective immunity. IMPORTANCE: Staphylococcus aureus is the leading cause of soft tissue and bloodstream infections; however, a vaccine with clinical efficacy is not available. Using mice to model staphylococcal infection, earlier work identified protective antigens; however, corresponding human clinical trials did not reach their endpoints. We show that B cell receptor (IgM) cross-linking by protein A is an important immune evasion strategy of S. aureus that can be monitored in a guinea pig model of bloodstream infection. Further, immunization with nontoxigenic protein A enables infected guinea pigs to elicit antibody responses that are protective against S. aureus. Thus, the guinea pig model may support preclinical development of staphylococcal vaccines.


Asunto(s)
Formación de Anticuerpos , Bacteriemia/inmunología , Infecciones Estafilocócicas/inmunología , Proteína Estafilocócica A/inmunología , Staphylococcus aureus/inmunología , Animales , Anticuerpos Antibacterianos/química , Anticuerpos Antibacterianos/inmunología , Bacteriemia/microbiología , Femenino , Cobayas , Humanos , Evasión Inmune , Inmunoglobulina M/química , Inmunoglobulina M/inmunología , Ratones , Ratones Endogámicos BALB C , Conejos , Infecciones Estafilocócicas/microbiología , Proteína Estafilocócica A/genética , Staphylococcus aureus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...