Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 291
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38876107

RESUMO

Vector-borne diseases are a leading cause of death worldwide and pose a substantial unmet medical need. Pathogens binding to host extracellular proteins (the "exoproteome") represents a crucial interface in the etiology of vector-borne disease. Here, we used bacterial selection to elucidate host-microbe interactions in high throughput (BASEHIT)-a technique enabling interrogation of microbial interactions with 3,324 human exoproteins-to profile the interactomes of 82 human-pathogen samples, including 30 strains of arthropod-borne pathogens and 8 strains of related non-vector-borne pathogens. The resulting atlas revealed 1,303 putative interactions, including hundreds of pairings with potential roles in pathogenesis, including cell invasion, tissue colonization, immune evasion, and host sensing. Subsequent functional investigations uncovered that Lyme disease spirochetes recognize epidermal growth factor as an environmental cue of transcriptional regulation and that conserved interactions between intracellular pathogens and thioredoxins facilitate cell invasion. In summary, this interactome atlas provides molecular-level insights into microbial pathogenesis and reveals potential host-directed targets for next-generation therapeutics.

2.
Trends Immunol ; 44(4): 256-265, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36964020

RESUMO

Malaria is caused by Plasmodium protozoa that are transmitted by anopheline mosquitoes. Plasmodium sporozoites are released with saliva when an infected female mosquito takes a blood meal on a vertebrate host. Sporozoites deposited into the skin must enter a blood vessel to start their journey towards the liver. After migration out of the mosquito, sporozoites are associated with, or in proximity to, many components of vector saliva in the skin. Recent work has elucidated how Anopheles saliva, and components of saliva, can influence host-pathogen interactions during the early stage of Plasmodium infection in the skin. Here, we discuss how components of Anopheles saliva can modulate local host responses and affect Plasmodium infectivity. We hypothesize that therapeutic strategies targeting mosquito salivary proteins can play a role in controlling malaria and other vector-borne diseases.


Assuntos
Anopheles , Malária , Humanos , Animais , Feminino , Anopheles/parasitologia , Anopheles/fisiologia , Saliva , Mosquitos Vetores/parasitologia , Esporozoítos
3.
PLoS Biol ; 21(10): e3002331, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37862360

RESUMO

Arthropod-borne pathogens cause some of the most important human and animal infectious diseases. Many vectors acquire or transmit pathogens through the process of blood feeding. Here, we report adiponectin, the most abundant adipocyte-derived hormone circulating in human blood, directly or indirectly inhibits acquisition of the Lyme disease agent, Borrelia burgdorferi, by Ixodes scapularis ticks. Rather than altering tick feeding or spirochete viability, adiponectin or its associated factors induces host histamine release when the tick feeds, which leads to vascular leakage, infiltration of neutrophils and macrophages, and inflammation at the bite site. Consistent with this, adiponectin-deficient mice have diminished pro-inflammatory responses, including interleukin (IL)-12 and IL-1ß, following a tick bite, compared with wild-type animals. All these factors mediated by adiponectin or associated factors influence B. burgdorferi survival at the tick bite site. These results suggest a host adipocyte-derived hormone modulates pathogen acquisition by a blood-feeding arthropod.


Assuntos
Grupo Borrelia Burgdorferi , Ixodes , Doença de Lyme , Picadas de Carrapatos , Animais , Camundongos , Humanos , Adiponectina , Grupo Borrelia Burgdorferi/fisiologia , Ixodes/fisiologia , Mamíferos
4.
Proc Natl Acad Sci U S A ; 120(20): e2208673120, 2023 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-37155900

RESUMO

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.


Assuntos
Artrópodes , Infecções Bacterianas , Borrelia burgdorferi , Ixodes , Doença de Lyme , Animais , Ixodes/microbiologia , Borrelia burgdorferi/genética , NF-kappa B , Doença de Lyme/microbiologia
5.
Nat Immunol ; 14(12): 1237-46, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24185615

RESUMO

Induction of type I interferon is a central event of innate immunity, essential for host defense. Here we report that the transcription factor ELF4 is induced by type I interferon and upregulates interferon expression in a feed-forward loop. ELF4 deficiency leads to reduced interferon production, resulting in enhanced susceptibility to West Nile virus encephalitis in mice. After viral infection, ELF4 is recruited by STING, interacts with and is activated by the MAVS-TBK1 complex, and translocates into the nucleus to bind interferon promoters. Cooperative binding with ELF4 increases the binding affinity of interferon regulatory factors IRF3 and IRF7, which is mediated by EICE elements. Thus, in addition to identifying a regulator of innate immune signaling, we uncovered a role for EICE elements in interferon transactivation.


Assuntos
Proteínas de Ligação a DNA/imunologia , Interferon beta/imunologia , Fatores de Transcrição/imunologia , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/imunologia , Animais , Linhagem Celular , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno/imunologia , Humanos , Immunoblotting , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/imunologia , Fator Regulador 7 de Interferon/metabolismo , Interferon beta/genética , Interferon beta/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Ligação Proteica/imunologia , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia , Análise de Sobrevida , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional/imunologia , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/fisiologia
6.
Cell ; 142(5): 714-25, 2010 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-20797779

RESUMO

West Nile virus (WNV) is the most common arthropod-borne flavivirus in the United States; however, the vector ligand(s) that participate in infection are not known. We now show that an Aedes aegypti C-type lectin, mosGCTL-1, is induced by WNV, interacts with WNV in a calcium-dependent manner, and facilitates infection in vivo and in vitro. A mosquito homolog of human CD45 in A. aegypti, designated mosPTP-1, recruits mosGCTL-1 to enable viral attachment to cells and to enhance viral entry. In vivo experiments show that mosGCTL-1 and mosPTP-1 function as part of the same pathway and are critical for WNV infection of mosquitoes. A similar phenomenon was also observed in Culex quinquefasciatus, a natural vector of WNV, further demonstrating that these genes participate in WNV infection. During the mosquito blood-feeding process, WNV infection was blocked in vivo with mosGCTL-1 antibodies. A molecular understanding of flaviviral-arthropod interactions may lead to strategies to control viral dissemination in nature.


Assuntos
Aedes/virologia , Culex/virologia , Proteínas de Insetos/metabolismo , Lectinas Tipo C/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Internalização do Vírus , Vírus do Nilo Ocidental/fisiologia , Animais , Humanos , Antígenos Comuns de Leucócito/química
7.
BMC Genomics ; 25(1): 42, 2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38191283

RESUMO

Gene-edited mosquitoes lacking a gamma-interferon-inducible lysosomal thiol reductase-like protein, namely (mosGILTnull) have lower Plasmodium infection, which is linked to impaired ovarian development and immune activation. The transcriptome of mosGILTnull Anopheles gambiae was therefore compared to wild type (WT) mosquitoes by RNA-sequencing to delineate mosGILT-dependent pathways. Compared to WT mosquitoes, mosGILTnull A. gambiae demonstrated altered expression of genes related to oogenesis, 20-hydroxyecdysone synthesis, as well as immune-related genes. Serendipitously, the zero population growth gene, zpg, an essential regulator of germ cell development was found to be one of the most downregulated genes in mosGILTnull mosquitoes. These results provide a crucial missing link between two previous studies on the role of zpg and mosGILT in ovarian development. This study further demonstrates that mosGILT has the potential to serve as a target for the biological control of mosquito vectors and to influence the Plasmodium life cycle within the vector.


Assuntos
Anopheles , Animais , Anopheles/genética , Diferenciação Celular , Imunidade Inata/genética , Mosquitos Vetores/genética , Células Germinativas
8.
Trends Genet ; 37(1): 8-11, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33020021

RESUMO

Ticks exist across diverse environments and transmit numerous pathogens. Due to their long and unique life cycles, these arthropods likely evolved robust epigenetic mechanisms that provide sustainable responses and buffers against extreme environmental conditions. Herein, we highlight how the study of the epigenetic basis of tick biology and vectorial capacity will enrich our knowledge of tick-borne infections.


Assuntos
Vetores Artrópodes/crescimento & desenvolvimento , Transmissão de Doença Infecciosa , Epigênese Genética , Interações Hospedeiro-Patógeno , Doenças Transmitidas por Carrapatos/epidemiologia , Carrapatos/fisiologia , Animais , Vetores Artrópodes/classificação , Vetores Artrópodes/genética , Humanos , Doenças Transmitidas por Carrapatos/genética , Doenças Transmitidas por Carrapatos/transmissão , Carrapatos/microbiologia , Carrapatos/parasitologia , Carrapatos/virologia
9.
Trends Immunol ; 42(7): 554-574, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34074602

RESUMO

Ticks have an unparalleled ability to parasitize diverse land vertebrates. Their natural persistence and vector competence are supported by the evolution of sophisticated hematophagy and remarkable host immune-evasion activities. We analyze the immunomodulatory roles of tick saliva which facilitates their acquisition of a blood meal from natural hosts and allows pathogen transmission. We also discuss the contrasting immunological events of tick-host associations in non-reservoir or incidental hosts, in which the development of acquired tick resistance can deter tick attachment. A critical appraisal of the intricate immunobiology of tick-host associations can plant new seeds of innovative research and contribute to the development of novel preventive strategies against ticks and tick-transmitted infections.


Assuntos
Carrapatos , Animais , Humanos , Evasão da Resposta Imune , Imunidade , Imunomodulação , Saliva
10.
Cell ; 139(7): 1243-54, 2009 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-20064371

RESUMO

Influenza viruses exploit host cell machinery to replicate, resulting in epidemics of respiratory illness. In turn, the host expresses antiviral restriction factors to defend against infection. To find host cell modifiers of influenza A H1N1 viral infection, we used a functional genomic screen and identified over 120 influenza A virus-dependency factors with roles in endosomal acidification, vesicular trafficking, mitochondrial metabolism, and RNA splicing. We discovered that the interferon-inducible transmembrane proteins IFITM1, 2, and 3 restrict an early step in influenza A viral replication. The IFITM proteins confer basal resistance to influenza A virus but are also inducible by interferons type I and II and are critical for interferon's virustatic actions. Further characterization revealed that the IFITM proteins inhibit the early replication of flaviviruses, including dengue virus and West Nile virus. Collectively this work identifies a family of antiviral restriction factors that mediate cellular innate immunity to at least three major human pathogens.


Assuntos
Infecções por Flavivirus/imunologia , Influenza Humana/imunologia , Proteínas de Membrana/imunologia , Animais , Antígenos de Diferenciação , Linhagem Celular Tumoral , Vírus da Dengue/imunologia , Humanos , Imunidade Inata , Vírus da Influenza A/imunologia , Interferons/imunologia , Camundongos , Proteínas de Ligação a RNA/imunologia , Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/fisiologia
11.
Mol Ther ; 31(9): 2702-2714, 2023 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-37533256

RESUMO

Lyme disease is the most common vector-borne infectious disease in the United States, in part because a vaccine against it is not currently available for humans. We propose utilizing the lipid nanoparticle-encapsulated nucleoside-modified mRNA (mRNA-LNP) platform to generate a Lyme disease vaccine like the successful clinical vaccines against SARS-CoV-2. Of the antigens expressed by Borrelia burgdorferi, the causative agent of Lyme disease, outer surface protein A (OspA) is the most promising candidate for vaccine development. We have designed and synthesized an OspA-encoding mRNA-LNP vaccine and compared its immunogenicity and protective efficacy to an alum-adjuvanted OspA protein subunit vaccine. OspA mRNA-LNP induced superior humoral and cell-mediated immune responses in mice after a single immunization. These potent immune responses resulted in protection against bacterial infection. Our study demonstrates that highly efficient mRNA vaccines can be developed against bacterial targets.


Assuntos
COVID-19 , Doença de Lyme , Humanos , Animais , Camundongos , Vacinas contra COVID-19 , COVID-19/prevenção & controle , SARS-CoV-2 , Doença de Lyme/prevenção & controle , Antígenos de Superfície/genética , Proteínas da Membrana Bacteriana Externa/genética
12.
Infect Immun ; 91(11): e0028223, 2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-37846980

RESUMO

Ticks are hematophagous arthropods that use a complex mixture of salivary proteins to evade host defenses while taking a blood meal. Little is known about the immunological and physiological consequences of tick feeding on humans. Here, we performed the first bulk and single-nucleus RNA sequencing (snRNA-seq) of skin and blood of four persons presenting with naturally acquired, attached Ixodes scapularis ticks. Pathways and individual genes associated with innate and adaptive immunity were identified based on bulk RNA sequencing, including interleukin-17 signaling and platelet activation pathways at the site of tick attachment or in peripheral blood. snRNA-seq further revealed that the Hippo signaling, cell adhesion, and axon guidance pathways were involved in the response to an I. scapularis bite in humans. Features of the host response in these individuals also overlapped with that of laboratory guinea pigs exposed to I. scapularis and which acquired resistance to ticks. These findings offer novel insights for the development of new biomarkers for I. scapularis exposure and anti-tick vaccines for human use.


Assuntos
Ixodes , Picadas de Carrapatos , Humanos , Animais , Cobaias , Ixodes/genética , Sequência de Bases , Comportamento Alimentar/fisiologia , RNA Nuclear Pequeno
13.
Nature ; 546(7660): 667-670, 2017 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-28636595

RESUMO

Rotavirus, a leading cause of severe gastroenteritis and diarrhoea in young children, accounts for around 215,000 deaths annually worldwide. Rotavirus specifically infects the intestinal epithelial cells in the host small intestine and has evolved strategies to antagonize interferon and NF-κB signalling, raising the question as to whether other host factors participate in antiviral responses in intestinal mucosa. The mechanism by which enteric viruses are sensed and restricted in vivo, especially by NOD-like receptor (NLR) inflammasomes, is largely unknown. Here we uncover and mechanistically characterize the NLR Nlrp9b that is specifically expressed in intestinal epithelial cells and restricts rotavirus infection. Our data show that, via RNA helicase Dhx9, Nlrp9b recognizes short double-stranded RNA stretches and forms inflammasome complexes with the adaptor proteins Asc and caspase-1 to promote the maturation of interleukin (Il)-18 and gasdermin D (Gsdmd)-induced pyroptosis. Conditional depletion of Nlrp9b or other inflammasome components in the intestine in vivo resulted in enhanced susceptibility of mice to rotavirus replication. Our study highlights an important innate immune signalling pathway that functions in intestinal epithelial cells and may present useful targets in the modulation of host defences against viral pathogens.


Assuntos
Células Epiteliais/imunologia , Células Epiteliais/virologia , Inflamassomos/metabolismo , Intestinos/citologia , Receptores Acoplados a Proteínas G/metabolismo , Infecções por Rotavirus/imunologia , Infecções por Rotavirus/virologia , Rotavirus/imunologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Caspase 1/metabolismo , RNA Helicases DEAD-box/metabolismo , Células Epiteliais/metabolismo , Feminino , Imunidade Inata , Inflamassomos/química , Inflamassomos/genética , Interleucina-18/imunologia , Mucosa Intestinal/metabolismo , Intestinos/imunologia , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Ligação a Fosfato , Piroptose , RNA de Cadeia Dupla/metabolismo , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/imunologia , Rotavirus/crescimento & desenvolvimento
14.
Infect Immun ; 90(1): e0035921, 2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-34724388

RESUMO

Malaria begins when an infected mosquito injects saliva containing Plasmodium sporozoites into the skin of a vertebrate host. Passive immunization of mice with antiserum against the Anopheles gambiae mosquito saliva protein TRIO (AgTRIO) offers significant protection against Plasmodium infection of mice. Furthermore, passive transfer of both AgTRIO antiserum and an anti-circumsporozoite protein monoclonal antibody provides synergistic protection. In this study, we generated monoclonal antibodies against AgTRIO to delineate the regions of AgTRIO associated with protective immunity. Monoclonal antibody 13F-1 markedly reduced Plasmodium infection in mice and recognized a region (VDDLMAKFN) in the carboxyl terminus of AgTRIO. 13F-1 is an IgG2a isotype monoclonal antibody, and the Fc region is required for protection. These data will aid in the generation of future malaria vaccines that may include both pathogen and vector antigens.


Assuntos
Anopheles/imunologia , Anticorpos Monoclonais/imunologia , Culicidae/imunologia , Malária/imunologia , Malária/prevenção & controle , Sequência de Aminoácidos , Animais , Modelos Animais de Doenças , Imunização Passiva , Fragmentos Fc das Imunoglobulinas , Proteínas de Insetos/química , Proteínas de Insetos/imunologia , Malária/parasitologia , Camundongos , Plasmodium berghei/imunologia , Ligação Proteica/imunologia , Domínios e Motivos de Interação entre Proteínas/imunologia
15.
PLoS Pathog ; 16(11): e1009030, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33175909

RESUMO

Lyme disease, the most common vector-borne illness in North America, is caused by the spirochete Borrelia burgdorferi. Infection begins in the skin following a tick bite and can spread to the hearts, joints, nervous system, and other organs. Diverse host responses influence the level of B. burgdorferi infection in mice and humans. Using a systems biology approach, we examined potential molecular interactions between human extracellular and secreted proteins and B. burgdorferi. A yeast display library expressing 1031 human extracellular proteins was probed against 36 isolates of B. burgdorferi sensu lato. We found that human Peptidoglycan Recognition Protein 1 (PGLYRP1) interacted with the vast majority of B. burgdorferi isolates. In subsequent experiments, we demonstrated that recombinant PGLYRP1 interacts with purified B. burgdorferi peptidoglycan and exhibits borreliacidal activity, suggesting that vertebrate hosts may use PGLYRP1 to identify B. burgdorferi. We examined B. burgdorferi infection in mice lacking PGLYRP1 and observed an increased spirochete burden in the heart and joints, along with splenomegaly. Mice lacking PGLYRP1 also showed signs of immune dysregulation, including lower serum IgG levels and higher levels of IFNγ, CXCL9, and CXCL10.Taken together, our findings suggest that PGLYRP1 plays a role in the host's response to B. burgdorferi and further demonstrate the utility of expansive yeast display screening in capturing biologically relevant interactions between spirochetes and their hosts.


Assuntos
Borrelia burgdorferi/fisiologia , Citocinas/metabolismo , Doença de Lyme/microbiologia , Animais , Citocinas/genética , Biblioteca Gênica , Humanos , Camundongos , Camundongos Endogâmicos BALB C
16.
Nat Immunol ; 11(10): 912-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20818395

RESUMO

Caspase-12 has been shown to negatively modulate inflammasome signaling during bacterial infection. Its function in viral immunity, however, has not been characterized. We now report an important role for caspase-12 in controlling viral infection via the pattern-recognition receptor RIG-I. After challenge with West Nile virus (WNV), caspase-12-deficient mice had greater mortality, higher viral burden and defective type I interferon response compared with those of challenged wild-type mice. In vitro studies of primary neurons and mouse embryonic fibroblasts showed that caspase-12 positively modulated the production of type I interferon by regulating E3 ubiquitin ligase TRIM25-mediated ubiquitination of RIG-I, a critical signaling event for the type I interferon response to WNV and other important viral pathogens.


Assuntos
Caspase 12/metabolismo , RNA Helicases DEAD-box/metabolismo , Interferon Tipo I/biossíntese , Receptores Virais/metabolismo , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental , Animais , Caspase 12/genética , Células Cultivadas , Proteína DEAD-box 58 , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/metabolismo , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Febre do Nilo Ocidental/genética
17.
J Infect Dis ; 224(3): 544-553, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-33306099

RESUMO

Malaria begins when Plasmodium-infected Anopheles mosquitoes take a blood meal on a vertebrate. During the initial probing process, mosquitoes inject saliva and sporozoites into the host skin. Components of mosquito saliva have the potential to influence sporozoite functionality. Sporozoite-associated mosquito saliva protein 1 (SAMSP1; AGAP013726) was among several proteins identified when sporozoites were isolated from saliva, suggesting it may have an effect on Plasmodium. Recombinant SAMSP1 enhanced sporozoite gliding and cell traversal activity in vitro. Moreover, SAMSP1 decreased neutrophil chemotaxis in vivo and in vitro, thereby also exerting an influence on the host environment in which the sporozoites reside. Active or passive immunization of mice with SAMSP1 or SAMSP1 antiserum diminished the initial Plasmodium burden after infection. Passive immunization of mice with SAMSP1 antiserum also added to the protective effect of a circumsporozoite protein monoclonal antibody. SAMSP1 is, therefore, a mosquito saliva protein that can influence sporozoite infectivity in the vertebrate host.


Assuntos
Anopheles , Plasmodium , Animais , Proteínas de Insetos , Malária , Camundongos , Proteínas de Protozoários , Proteínas e Peptídeos Salivares , Esporozoítos
18.
J Infect Dis ; 223(12): 2186-2196, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-33161431

RESUMO

The stimulator of interferon gene (STING) pathway controls both DNA and RNA virus infection. STING is essential for induction of innate immune responses during DNA virus infection, while its mechanism against RNA virus remains largely elusive. We show that STING signaling is crucial for restricting chikungunya virus infection and arthritis pathogenesis. Sting-deficient mice (Stinggt/gt) had elevated viremia throughout the viremic stage and viral burden in feet transiently, with a normal type I IFN response. Stinggt/gt mice presented much greater foot swelling, joint damage, and immune cell infiltration than wild-type mice. Intriguingly, expression of interferon-γ and Cxcl10 was continuously upregulated by approximately 7 to 10-fold and further elevated in Stinggt/gt mice synchronously with arthritis progression. However, expression of chemoattractants for and activators of neutrophils, Cxcl5, Cxcl7, and Cxcr2 was suppressed in Stinggt/gt joints. These results demonstrate that STING deficiency leads to an aberrant chemokine response that promotes pathogenesis of CHIKV arthritis.


Assuntos
Artrite , Febre de Chikungunya , Proteínas de Membrana/imunologia , Animais , Artrite/imunologia , Artrite/virologia , Febre de Chikungunya/imunologia , Vírus Chikungunya , Imunidade Inata , Camundongos , Camundongos Knockout , Viremia
19.
Curr Issues Mol Biol ; 42: 113-144, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33289683

RESUMO

Borrelia burgdorferi sensu lato causes Lyme borreliosis in a variety of animals and humans. These atypical bacterial pathogens are maintained in a complex enzootic life cycle that primarily involves a vertebrate host and Ixodes spp. ticks. In the Northeastern United States, I. scapularis is the main vector, while wild rodents serve as the mammalian reservoir host. As B. burgdorferi is transmitted only by I. scapularis and closely related ticks, the spirochete-tick interactions are thought to be highly specific. Various borrelial and arthropod proteins that directly or indirectly contribute to the natural cycle of B. burgdorferi infection have been identified. Discrete molecular interactions between spirochetes and tick components also have been discovered, which often play critical roles in pathogen persistence and transmission by the arthropod vector. This review will focus on the past discoveries and future challenges that are relevant to our understanding of the molecular interactions between B. burgdorferi and Ixodes ticks. This information will not only impact scientific advancements in the research of tick- transmitted infections but will also contribute to the development of novel preventive measures that interfere with the B. burgdorferi life cycle.


Assuntos
Vetores Aracnídeos/microbiologia , Borrelia burgdorferi , Interações Hospedeiro-Patógeno , Doença de Lyme/microbiologia , Doença de Lyme/transmissão , Carrapatos/microbiologia , Animais , Vetores Aracnídeos/crescimento & desenvolvimento , Humanos , Ixodes/microbiologia , Estágios do Ciclo de Vida , Doença de Lyme/epidemiologia , Doença de Lyme/prevenção & controle , Noroeste dos Estados Unidos/epidemiologia , Carrapatos/crescimento & desenvolvimento
20.
Trends Immunol ; 39(11): 862-873, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30301592

RESUMO

Recent scientific breakthroughs have significantly expanded our understanding of arthropod vector immunity. Insights in the laboratory have demonstrated how the immune system provides resistance to infection, and in what manner innate defenses protect against a microbial assault. Less understood, however, is the effect of biotic and abiotic factors on microbial-vector interactions and the impact of the immune system on arthropod populations in nature. Furthermore, the influence of genetic plasticity on the immune response against vector-borne pathogens remains mostly elusive. Herein, we discuss evolutionary forces that shape arthropod vector immunity. We focus on resistance, pathogenicity and tolerance to infection. We posit that novel scientific paradigms should emerge when molecular immunologists and evolutionary ecologists work together.


Assuntos
Vetores Artrópodes/imunologia , Artrópodes/imunologia , Mamíferos/imunologia , Animais , Evolução Biológica , Ecologia , Humanos , Tolerância Imunológica , Imunidade , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA