Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Cell ; 180(2): 216-218, 2020 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-31978340

RESUMO

Microbes that cause persistent infections (e.g., herpes viruses) do so by switching from fast-growing lytic states to slow-growing latent states. Waldman et al. have identified a single transcription factor that governs the switch between the lytic and latent forms of Toxoplasma gondii, a parasite that causes a persistent brain infection.


Assuntos
Toxoplasma , Encéfalo , Diferenciação Celular , Regulação da Expressão Gênica , Fatores de Transcrição
2.
PLoS Pathog ; 19(4): e1011347, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37068104

RESUMO

Toxoplasma gondii establishes a long-lived latent infection in the central nervous system (CNS) of its hosts. Reactivation in immunocompromised individuals can lead to life threatening disease. Latent infection is driven by the ability of the parasite to convert from the acute-stage tachyzoite to the latent-stage bradyzoite which resides in long-lived intracellular cysts. While much work has focused on the parasitic factors that drive cyst development, the host factors that influence encystment are not well defined. Here we show that a polymorphic secreted parasite kinase (ROP16), that phosphorylates host cell proteins, mediates efficient encystment of T. gondii in a stress-induced model of encystment and primary neuronal cell cultures (PNCs) in a strain-specific manner. Using short-hairpin RNA (shRNA) knockdowns in human foreskin fibroblasts (HFFs) and PNCs from transgenic mice, we determined that ROP16's cyst enhancing abilities are mediated, in part, by phosphorylation-and therefore activation-of the host cell transcription factor STAT6. To test the role of STAT6 in vivo, we infected wild-type (WT) and STAT6KO mice, finding that, compared to WT mice, STAT6KO mice have a decrease in CNS cyst burden but not overall parasite burden or dissemination to the CNS. Finally, we found a similar ROP16-dependent encystment defect in human pluripotent stem cell-derived neurons. Together, these findings identify a host cell factor (STAT6) that T. gondii manipulates in a strain-specific manner to generate a favorable encystment environment.


Assuntos
Toxoplasma , Camundongos , Animais , Humanos , Toxoplasma/fisiologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Fosforilação , Sistema Nervoso Central/metabolismo , Regulação da Expressão Gênica , Fator de Transcrição STAT6/metabolismo
3.
PLoS Pathog ; 18(6): e1010296, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35727849

RESUMO

Initial TCR engagement (priming) of naive CD8+ T cells results in T cell expansion, and these early events influence the generation of diverse effector and memory populations. During infection, activated T cells can re-encounter cognate antigen, but how these events influence local effector responses or formation of memory populations is unclear. To address this issue, OT-I T cells which express the Nur77-GFP reporter of TCR activation were paired with the parasite Toxoplasma gondii that expresses OVA to assess how secondary encounter with antigen influences CD8+ T cell responses. During acute infection, TCR stimulation in affected tissues correlated with parasite burden and was associated with markers of effector cells while Nur77-GFP- OT-I showed signs of effector memory potential. However, both Nur77-GFP- and Nur77-GFP+ OT-I from acutely infected mice formed similar memory populations when transferred into naive mice. During the chronic stage of infection in the CNS, TCR activation was associated with large scale transcriptional changes and the acquisition of an effector T cell phenotype as well as the generation of a population of CD103+ CD69+ Trm like cells. While inhibition of parasite replication resulted in reduced effector responses it did not alter the Trm population. These data sets highlight that recent TCR activation contributes to the phenotypic heterogeneity of the CD8+ T cell response but suggest that this process has a limited impact on memory populations at acute and chronic stages of infection.


Assuntos
Toxoplasma , Toxoplasmose , Animais , Linfócitos T CD8-Positivos , Memória Imunológica , Camundongos , Receptores de Antígenos de Linfócitos T
4.
PLoS Pathog ; 15(10): e1007856, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31648279

RESUMO

Toxoplasma gondii is an intracellular parasite that persistently infects the CNS and that has genetically distinct strains which provoke different acute immune responses. How differences in the acute immune response affect the CNS immune response is unknown. To address this question, we used two persistent Toxoplasma strains (type II and type III) and examined the CNS immune response at 21 days post infection (dpi). Contrary to acute infection studies, type III-infected mice had higher numbers of total CNS T cells and macrophages/microglia but fewer alternatively activated macrophages (M2s) and regulatory T cells (Tregs) than type II-infected mice. By profiling splenocytes at 5, 10, and 21 dpi, we determined that at 5 dpi type III-infected mice had more M2s while type II-infected mice had more pro-inflammatory macrophages and that these responses flipped over time. To test how these early differences influence the CNS immune response, we engineered the type III strain to lack ROP16 (IIIΔrop16), the polymorphic effector protein that drives the early type III-associated M2 response. IIIΔrop16-infected mice showed a type II-like neuroinflammatory response with fewer infiltrating T cells and macrophages/microglia and more M2s and an unexpectedly low CNS parasite burden. At 5 dpi, IIIΔrop16-infected mice showed a mixed inflammatory response with more pro-inflammatory macrophages, M2s, T effector cells, and Tregs, and decreased rates of infection of peritoneal exudative cells (PECs). These data suggested that type III parasites need the early ROP16-associated M2 response to avoid clearance, possibly by the Immunity-Related GTPases (IRGs), which are IFN-γ- dependent proteins essential for murine defenses against Toxoplasma. To test this possibility, we infected IRG-deficient mice and found that IIIΔrop16 parasites now maintained parental levels of PECs infection. Collectively, these studies suggest that, for the type III strain, rop16III plays a key role in parasite persistence and influences the subacute CNS immune response.


Assuntos
Sistema Nervoso Central/imunologia , Macrófagos/imunologia , Proteínas Tirosina Quinases/imunologia , Proteínas de Protozoários/imunologia , Linfócitos T/imunologia , Toxoplasma/imunologia , Toxoplasmose Animal/imunologia , Animais , Sistema Nervoso Central/parasitologia , GTP Fosfo-Hidrolases/genética , Camundongos , Camundongos Knockout , Microglia/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas de Protozoários/genética , Toxoplasma/classificação , Toxoplasma/genética
5.
PLoS Pathog ; 13(7): e1006351, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28727854

RESUMO

Toxoplasma gondii is one of the world's most successful parasites, in part because of its ability to infect and persist in most warm-blooded animals. A unique characteristic of T. gondii is its ability to persist in the central nervous system (CNS) of a variety of hosts, including humans and rodents. How, what, and why T. gondii encysts in the CNS has been the topic of study for decades. In this review, we will discuss recent work on how T. gondii is able to traverse the unique barrier surrounding the CNS, what cells of the CNS play host to T. gondii, and finally, how T. gondii infection may influence global and cellular physiology of the CNS.


Assuntos
Sistema Nervoso Central/parasitologia , Toxoplasma/fisiologia , Toxoplasmose/parasitologia , Animais , Sistema Nervoso Central/fisiologia , Humanos , Toxoplasma/genética , Toxoplasmose/fisiopatologia
6.
PLoS Pathog ; 12(2): e1005447, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26895155

RESUMO

Toxoplasma gondii, a common brain-tropic parasite, is capable of infecting most nucleated cells, including astrocytes and neurons, in vitro. Yet, in vivo, Toxoplasma is primarily found in neurons. In vitro data showing that interferon-γ-stimulated astrocytes, but not neurons, clear intracellular parasites suggest that neurons alone are persistently infected in vivo because they lack the ability to clear intracellular parasites. Here we test this theory by using a novel Toxoplasma-mouse model capable of marking and tracking host cells that directly interact with parasites, even if the interaction is transient. Remarkably, we find that Toxoplasma shows a strong predilection for interacting with neurons throughout CNS infection. This predilection remains in the setting of IFN-γ depletion; infection with parasites resistant to the major mechanism by which murine astrocytes clear parasites; or when directly injecting parasites into the brain. These findings, in combination with prior work, strongly suggest that neurons are not incidentally infected, but rather they are Toxoplasma's primary in vivo target.


Assuntos
Astrócitos/parasitologia , Encéfalo/parasitologia , Neurônios/parasitologia , Toxoplasma , Toxoplasmose/parasitologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Interferon gama/metabolismo , Espaço Intracelular/parasitologia , Camundongos
7.
PLoS Pathog ; 10(4): e1004047, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24722202

RESUMO

During infection with the intracellular parasite Toxoplasma gondii, the presentation of parasite-derived antigens to CD4+ and CD8+ T cells is essential for long-term resistance to this pathogen. Fundamental questions remain regarding the roles of phagocytosis and active invasion in the events that lead to the processing and presentation of parasite antigens. To understand the most proximal events in this process, an attenuated non-replicating strain of T. gondii (the cpsII strain) was combined with a cytometry-based approach to distinguish active invasion from phagocytic uptake. In vivo studies revealed that T. gondii disproportionately infected dendritic cells and macrophages, and that infected dendritic cells and macrophages displayed an activated phenotype characterized by enhanced levels of CD86 compared to cells that had phagocytosed the parasite, thus suggesting a role for these cells in priming naïve T cells. Indeed, dendritic cells were required for optimal CD4+ and CD8+ T cell responses, and the phagocytosis of heat-killed or invasion-blocked parasites was not sufficient to induce T cell responses. Rather, the selective transfer of cpsII-infected dendritic cells or macrophages (but not those that had phagocytosed the parasite) to naïve mice potently induced CD4+ and CD8+ T cell responses, and conferred protection against challenge with virulent T. gondii. Collectively, these results point toward a critical role for actively infected host cells in initiating T. gondii-specific CD4+ and CD8+ T cell responses.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Imunidade Celular , Toxoplasma/imunologia , Toxoplasmose/imunologia , Animais , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/patologia , Células Dendríticas/patologia , Camundongos , Toxoplasmose/genética , Toxoplasmose/patologia
8.
J Immunol ; 193(1): 139-49, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24860191

RESUMO

The balance between controlling infection and limiting inflammation is particularly precarious in the brain because of its unique vulnerability to the toxic effects of inflammation. Astrocytes have been implicated as key regulators of neuroinflammation in CNS infections, including infection with Toxoplasma gondii, a protozoan parasite that naturally establishes a chronic CNS infection in mice and humans. In CNS toxoplasmosis, astrocytes are critical to controlling parasite growth. They secrete proinflammatory cytokines and physically encircle parasites. However, the molecular mechanisms used by astrocytes to limit neuroinflammation during toxoplasmic encephalitis have not yet been identified. TGF-ß signaling in astrocytes is of particular interest because TGF-ß is universally upregulated during CNS infection and serves master regulatory and primarily anti-inflammatory functions. We report in this study that TGF-ß signaling is activated in astrocytes during toxoplasmic encephalitis and that inhibition of astrocytic TGF-ß signaling increases immune cell infiltration, uncouples proinflammatory cytokine and chemokine production from CNS parasite burden, and increases neuronal injury. Remarkably, we show that the effects of inhibiting astrocytic TGF-ß signaling are independent of parasite burden and the ability of GFAP(+) astrocytes to physically encircle parasites.


Assuntos
Astrócitos/imunologia , Neurônios/imunologia , Transdução de Sinais/imunologia , Toxoplasma/imunologia , Toxoplasmose Cerebral/imunologia , Fator de Crescimento Transformador beta/imunologia , Animais , Astrócitos/parasitologia , Astrócitos/patologia , Quimiocinas/genética , Quimiocinas/imunologia , Proteína Glial Fibrilar Ácida , Humanos , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/imunologia , Neurônios/parasitologia , Neurônios/patologia , Transdução de Sinais/genética , Toxoplasma/genética , Toxoplasmose Cerebral/genética , Toxoplasmose Cerebral/patologia , Fator de Crescimento Transformador beta/genética , Regulação para Cima/genética , Regulação para Cima/imunologia
9.
Proc Natl Acad Sci U S A ; 110(21): E1913-22, 2013 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-23650399

RESUMO

Toxoplasma gondii infection occurs through the oral route, but we lack important information about how the parasite interacts with the host immune system in the intestine. We used two-photon laser-scanning microscopy in conjunction with a mouse model of oral T. gondii infection to address this issue. T. gondii established discrete foci of infection in the small intestine, eliciting the recruitment and transepithelial migration of neutrophils and inflammatory monocytes. Neutrophils accounted for a high proportion of actively invaded cells, and we provide evidence for a role for transmigrating neutrophils and other immune cells in the spread of T. gondii infection through the lumen of the intestine. Our data identify neutrophils as motile reservoirs of T. gondii infection and suggest a surprising retrograde pathway for parasite spread in the intestine.


Assuntos
Movimento Celular/imunologia , Intestino Delgado/imunologia , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Toxoplasma/imunologia , Toxoplasmose/imunologia , Animais , Modelos Animais de Doenças , Imunidade Inata , Mucosa Intestinal/imunologia , Mucosa Intestinal/parasitologia , Mucosa Intestinal/patologia , Intestino Delgado/parasitologia , Intestino Delgado/patologia , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Neutrófilos/parasitologia , Neutrófilos/patologia , Toxoplasmose/parasitologia , Toxoplasmose/patologia
10.
PLoS Pathog ; 9(5): e1003331, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23658519

RESUMO

Toxoplasma gondii is an intracellular parasite that transitions from acute infection to a chronic infective state in its intermediate host via encystation, which enables the parasite to evade immune detection and clearance. It is widely accepted that the tissue cyst perimeter is highly and specifically decorated with glycan modifications; however, the role of these modifications in the establishment and persistence of chronic infection has not been investigated. Here we identify and biochemically and biologically characterize a Toxoplasma nucleotide-sugar transporter (TgNST1) that is required for cyst wall glycosylation. Toxoplasma strains deleted for the TgNST1 gene (Δnst1) form cyst-like structures in vitro but no longer interact with lectins, suggesting that Δnst1 strains are deficient in the transport and use of sugars for the biosynthesis of cyst-wall structures. In vivo infection experiments demonstrate that the lack of TgNST1 activity does not detectably impact the acute (tachyzoite) stages of an infection or tropism of the parasite for the brain but that Δnst1 parasites are severely defective in persistence during the chronic stages of the infection. These results demonstrate for the first time the critical role of parasite glycoconjugates in the persistence of Toxoplasma tissue cysts.


Assuntos
Glicoproteínas/metabolismo , Proteínas de Transporte de Nucleotídeos/metabolismo , Proteínas de Protozoários/metabolismo , Toxoplasma/metabolismo , Toxoplasmose/metabolismo , Animais , Feminino , Deleção de Genes , Glicoproteínas/genética , Glicosilação , Camundongos , Proteínas de Transporte de Nucleotídeos/genética , Proteínas de Protozoários/genética , Toxoplasma/genética , Toxoplasmose/genética , Toxoplasmose/patologia
11.
Eukaryot Cell ; 13(8): 965-76, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24951442

RESUMO

Intracellular pathogens can replicate efficiently only after they manipulate and modify their host cells to create an environment conducive to replication. While diverse cellular pathways are targeted by different pathogens, metabolism, membrane and cytoskeletal architecture formation, and cell death are the three primary cellular processes that are modified by infections. Toxoplasma gondii is an obligate intracellular protozoan that infects ∼30% of the world's population and causes severe and life-threatening disease in developing fetuses, in immune-comprised patients, and in certain otherwise healthy individuals who are primarily found in South America. The high prevalence of Toxoplasma in humans is in large part a result of its ability to modulate these three host cell processes. Here, we highlight recent work defining the mechanisms by which Toxoplasma interacts with these processes. In addition, we hypothesize why some processes are modified not only in the infected host cell but also in neighboring uninfected cells.


Assuntos
Toxoplasma/fisiologia , Toxoplasmose/imunologia , Animais , Apoptose , Metabolismo dos Carboidratos , Membrana Celular/metabolismo , Membrana Celular/parasitologia , Núcleo Celular/parasitologia , Núcleo Celular/fisiologia , Metabolismo Energético , Interações Hospedeiro-Parasita , Humanos , Inflamassomos/fisiologia , Metabolismo dos Lipídeos , Toxoplasmose/parasitologia
12.
Infect Immun ; 82(10): 4056-67, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25024368

RESUMO

The intracellular parasite Toxoplasma gondii has multiple strategies to alter host cell function, including the injection of rhoptry proteins into the cytosol of host cells as well as bystander populations, but the consequence of these events is unclear. Here, a reporter system using fluorescent parasite strains that inject Cre recombinase with their rhoptry proteins (Toxoplasma-Cre) was combined with Ai6 Cre reporter mice to identify cells that have been productively infected, that have been rhoptry injected but lack the parasite, or that have phagocytosed T. gondii. The ability to distinguish these host-parasite interactions was then utilized to dissect the events that lead to the production of interleukin-12 p40 (IL-12p40), which is required for resistance to T. gondii. In vivo, the use of invasion-competent or invasion-inhibited (phagocytosed) parasites with IL-12p40 (YET40) reporter mice revealed that dendritic cell (DC) and macrophage populations that phagocytose the parasite or are infected can express IL-12p40 but are not the major source, as larger numbers of uninfected cells secrete this cytokine. Similarly, the use of Toxoplasma-Cre parasite strains indicated that dendritic cells and inflammatory monocytes untouched by the parasite and not cells injected by the parasite are the primary source of IL-12p40. These results imply that a soluble host or parasite factor is responsible for the bulk of IL-12p40 production in vivo, rather than cellular interactions with T. gondii that result in infection, infection and clearance, injection of rhoptry proteins, or phagocytosis of the parasite.


Assuntos
Interações Hospedeiro-Parasita , Subunidade p40 da Interleucina-12/imunologia , Toxoplasma/genética , Toxoplasma/imunologia , Toxoplasmose/imunologia , Toxoplasmose/parasitologia , Animais , Células Dendríticas/imunologia , Células Dendríticas/parasitologia , Macrófagos/imunologia , Macrófagos/parasitologia , Camundongos , Monócitos/imunologia , Monócitos/parasitologia
13.
Immunol Cell Biol ; 92(10): 872-81, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25155465

RESUMO

The classic anti-viral cytokine interferon (IFN)-ß can be induced during parasitic infection, but relatively little is know about the cell types and signaling pathways involved. Here we show that inflammatory monocytes (IMs), but not neutrophils, produce IFN-ß in response to T. gondii infection. This difference correlated with the mode of parasite entry into host cells, with phagocytic uptake predominating in IMs and active invasion predominating in neutrophils. We also show that expression of IFN-ß requires phagocytic uptake of the parasite by IMs, and signaling through Toll-like receptors (TLRs) and MyD88. Finally, we show that IMs are major producers of IFN-ß in mesenteric lymph nodes following in vivo oral infection of mice, and mice lacking the receptor for type I IFN-1 show higher parasite loads and reduced survival. Our data reveal a TLR and internalization-dependent pathway in IMs for IFN-ß induction to a non-viral pathogen.


Assuntos
Interferon beta/biossíntese , Monócitos/imunologia , Receptores Toll-Like/metabolismo , Toxoplasmose Animal/imunologia , Animais , Imunidade Inata , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo , Neutrófilos/imunologia , Transdução de Sinais , Toxoplasma/imunologia , Toxoplasmose Animal/parasitologia
14.
PLoS Pathog ; 8(7): e1002825, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22910631

RESUMO

Like many intracellular microbes, the protozoan parasite Toxoplasma gondii injects effector proteins into cells it invades. One group of these effector proteins is injected from specialized organelles called the rhoptries, which have previously been described to discharge their contents only during successful invasion of a host cell. In this report, using several reporter systems, we show that in vitro the parasite injects rhoptry proteins into cells it does not productively invade and that the rhoptry effector proteins can manipulate the uninfected cell in a similar manner to infected cells. In addition, as one of the reporter systems uses a rhoptry:Cre recombinase fusion protein, we show that in Cre-reporter mice infected with an encysting Toxoplasma-Cre strain, uninfected-injected cells, which could be derived from aborted invasion or cell-intrinsic killing after invasion, are actually more common than infected-injected cells, especially in the mouse brain, where Toxoplasma encysts and persists. This phenomenon has important implications for how Toxoplasma globally affects its host and opens a new avenue for how other intracellular microbes may similarly manipulate the host environment at large.


Assuntos
Fibroblastos/parasitologia , Interações Hospedeiro-Parasita , Proteínas Tirosina Quinases/metabolismo , Proteínas de Protozoários/metabolismo , Toxoplasma/metabolismo , Toxoplasma/patogenicidade , Animais , Linhagem Celular , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição STAT6/metabolismo
15.
PLoS One ; 19(3): e0300764, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38551902

RESUMO

Toxoplasma gondii is an intracellular parasite that establishes a long-term infection in the brain of many warm-blooded hosts, including humans and rodents. Like all obligate intracellular microbes, Toxoplasma uses many effector proteins to manipulate the host cell to ensure parasite survival. While some of these effector proteins are universal to all Toxoplasma strains, some are polymorphic between Toxoplasma strains. One such polymorphic effector is GRA15. The gra15 allele carried by type II strains activates host NF-κB signaling, leading to the release of cytokines such as IL-12, TNF, and IL-1ß from immune cells infected with type II parasites. Prior work also suggested that GRA15 promotes early host control of parasites in vivo, but the effect of GRA15 on parasite persistence in the brain and the peripheral immune response has not been well defined. For this reason, we sought to address this gap by generating a new IIΔgra15 strain and comparing outcomes at 3 weeks post infection between WT and IIΔgra15 infected mice. We found that the brain parasite burden and the number of macrophages/microglia and T cells in the brain did not differ between WT and IIΔgra15 infected mice. In addition, while IIΔgra15 infected mice had a lower number and frequency of splenic M1-like macrophages and frequency of PD-1+ CTLA-4+ CD4+ T cells and NK cells compared to WT infected mice, the IFN-γ+ CD4 and CD8 T cell populations were equivalent. In summary, our results suggest that in vivo GRA15 may have a subtle effect on the peripheral immune response, but this effect is not strong enough to alter brain parasite burden or parenchymal immune cell number at 3 weeks post infection.


Assuntos
Toxoplasma , Humanos , Animais , Camundongos , Proteínas de Protozoários/metabolismo , Transdução de Sinais , Citocinas/metabolismo , NF-kappa B/metabolismo
16.
Nat Microbiol ; 9(8): 2051-2072, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39075233

RESUMO

Delivering macromolecules across biological barriers such as the blood-brain barrier limits their application in vivo. Previous work has demonstrated that Toxoplasma gondii, a parasite that naturally travels from the human gut to the central nervous system (CNS), can deliver proteins to host cells. Here we engineered T. gondii's endogenous secretion systems, the rhoptries and dense granules, to deliver multiple large (>100 kDa) therapeutic proteins into neurons via translational fusions to toxofilin and GRA16. We demonstrate delivery in cultured cells, brain organoids and in vivo, and probe protein activity using imaging, pull-down assays, scRNA-seq and fluorescent reporters. We demonstrate robust delivery after intraperitoneal administration in mice and characterize 3D distribution throughout the brain. As proof of concept, we demonstrate GRA16-mediated brain delivery of the MeCP2 protein, a putative therapeutic target for Rett syndrome. By characterizing the potential and current limitations of the system, we aim to guide future improvements that will be required for broader application.


Assuntos
Encéfalo , Neurônios , Proteínas de Protozoários , Toxoplasma , Toxoplasma/genética , Toxoplasma/metabolismo , Animais , Neurônios/metabolismo , Neurônios/parasitologia , Camundongos , Humanos , Encéfalo/metabolismo , Encéfalo/parasitologia , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/genética , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Sistemas de Liberação de Medicamentos
17.
Nat Methods ; 7(4): 307-9, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20208532

RESUMO

We describe a Toxoplasma gondii strain that will permit the use of site-specific recombination to study the host-parasite interactions of this organism. This Toxoplasma strain efficiently injects a Cre fusion protein into host cells. In a Cre-reporter cell line, a single parasite invasion induced Cre-mediated recombination in 95% of infected host cells. By infecting Cre-reporter mice with these parasites, we also monitored host-cell infection in vivo.


Assuntos
Integrases/metabolismo , Toxoplasma/enzimologia , Toxoplasmose/parasitologia , Animais , Interações Hospedeiro-Parasita , Integrases/genética , Integrases/imunologia , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Plasmídeos/genética , Recombinação Genética , Toxoplasma/genética , Transdução Genética
18.
Cell Host Microbe ; 31(10): 1748-1762.e8, 2023 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-37827122

RESUMO

Intracellular pathogens and other endosymbionts reprogram host cell transcription to suppress immune responses and recalibrate biosynthetic pathways. This reprogramming is critical in determining the outcome of infection or colonization. We combine pooled CRISPR knockout screening with dual host-microbe single-cell RNA sequencing, a method we term dual perturb-seq, to identify the molecular mediators of these transcriptional interactions. Applying dual perturb-seq to the intracellular pathogen Toxoplasma gondii, we are able to identify previously uncharacterized effector proteins and directly infer their function from the transcriptomic data. We show that TgGRA59 contributes to the export of other effector proteins from the parasite into the host cell and identify an effector, TgSOS1, that is necessary for sustained host STAT6 signaling and thereby contributes to parasite immune evasion and persistence. Together, this work demonstrates a tool that can be broadly adapted to interrogate host-microbe transcriptional interactions and reveal mechanisms of infection and immune evasion.


Assuntos
Toxoplasma , Toxoplasma/genética , Perfilação da Expressão Gênica , Transcriptoma , Evasão da Resposta Imune , Transdução de Sinais , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo
19.
Nat Microbiol ; 8(5): 889-904, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37081202

RESUMO

Successful infection strategies must balance pathogen amplification and persistence. In the obligate intracellular parasite Toxoplasma gondii this is accomplished through differentiation into dedicated cyst-forming chronic stages that avoid clearance by the host immune system. The transcription factor BFD1 is both necessary and sufficient for stage conversion; however, its regulation is not understood. In this study we examine five factors that are transcriptionally activated by BFD1. One of these is a cytosolic RNA-binding protein of the CCCH-type zinc-finger family, which we name bradyzoite formation deficient 2 (BFD2). Parasites lacking BFD2 fail to induce BFD1 and are consequently unable to fully differentiate in culture or in mice. BFD2 interacts with the BFD1 transcript under stress, and deletion of BFD2 reduces BFD1 protein levels but not messenger RNA abundance. The reciprocal effects on BFD2 transcription and BFD1 translation outline a positive feedback loop that enforces the chronic-stage gene-expression programme. Thus, our findings help explain how parasites both initiate and commit to chronic differentiation. This work provides new mechanistic insight into the regulation of T. gondii persistence, and can be exploited in the design of strategies to prevent and treat these key reservoirs of human infection.


Assuntos
Toxoplasma , Camundongos , Animais , Humanos , Toxoplasma/metabolismo , Retroalimentação , Regulação da Expressão Gênica , Fatores de Transcrição/genética
20.
Trends Parasitol ; 38(12): 1026-1027, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36302693

RESUMO

Using a CRISPR/Cas9-based method, Wincott et al. generated a stable, complex Toxoplasma gondii population composed of 96 barcoded clonal lineages. By tracking the population structure in vivo, they determine that - contrary to expectations - the pathway to infecting the brain is widely permissive for T. gondii.


Assuntos
Parasitos , Toxoplasma , Animais , Toxoplasma/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA