Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
SLAS Discov ; 26(4): 534-546, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33445986

RESUMO

Targeted protein degradation represents an area of great interest, potentially offering improvements with respect to dosing, side effects, drug resistance, and reaching "undruggable" proteins compared with traditional small-molecule therapeutics. A major challenge in the design and characterization of degraders acting as molecular glues is that binding of the molecule to the protein of interest (PoI) is not needed for efficient and selective protein degradation; instead, one needs to understand the interaction with the responsible ligase. Similarly, for proteasome targeting chimeras (PROTACs), understanding the binding characteristics of the PoI alone is not sufficient. Therefore, simultaneously assessing the binding to both PoI and the E3 ligase as well as the resulting degradation profile is of great value. The cellular thermal shift assay (CETSA) is an unbiased cell-based method, designed to investigate the interaction of compounds with their cellular protein targets by measuring compound-induced changes in protein thermal stability. In combination with mass spectrometry (MS), CETSA can simultaneously evaluate compound-induced changes in the stability of thousands of proteins. We have used CETSA MS to profile a number of protein degraders, including molecular glues (e.g., immunomodulatory drugs) and PROTACs, to understand mode of action and to deconvolute off-target effects in intact cells. Within the same experiment, we were able to monitor both target engagement by observing changes in protein thermal stability as well as efficacy by simultaneous assessment of protein abundances. This allowed us to correlate target engagement (i.e., binding to the PoI and ligases) and functional readout (i.e., degrader induced protein degradation).


Assuntos
Ensaios de Triagem em Larga Escala , Agentes de Imunomodulação/farmacologia , Terapia de Alvo Molecular/métodos , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional , Ubiquitina-Proteína Ligases/metabolismo , Descoberta de Drogas/métodos , Células Eucarióticas/citologia , Células Eucarióticas/efeitos dos fármacos , Células Eucarióticas/imunologia , Células Eucarióticas/metabolismo , Humanos , Agentes de Imunomodulação/química , Ligantes , Espectrometria de Massas/métodos , Ligação Proteica , Estabilidade Proteica , Proteólise/efeitos dos fármacos , Proteômica/métodos , Proteostase/genética , Temperatura , Ubiquitina-Proteína Ligases/genética , Ubiquitinação/efeitos dos fármacos
2.
Annu Rev Pathol ; 15: 439-466, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31657966

RESUMO

Legionella species are environmental gram-negative bacteria able to cause a severe form of pneumonia in humans known as Legionnaires' disease. Since the identification of Legionella pneumophila in 1977, four decades of research on Legionella biology and Legionnaires' disease have brought important insights into the biology of the bacteria and the molecular mechanisms that these intracellular pathogens use to cause disease in humans. Nowadays, Legionella species constitute a remarkable model of bacterial adaptation, with a genus genome shaped by their close coevolution with amoebae and an ability to exploit many hosts and signaling pathways through the secretion of a myriad of effector proteins, many of which have a eukaryotic origin. This review aims to discuss current knowledge of Legionella infection mechanisms and future research directions to be taken that might answer the many remaining open questions. This research will without a doubt be a terrific scientific journey worth taking.


Assuntos
Legionella pneumophila/patogenicidade , Doença dos Legionários/microbiologia , Adaptação Fisiológica/genética , Adaptação Fisiológica/imunologia , Amoeba/genética , Amoeba/imunologia , Amoeba/patogenicidade , Células Eucarióticas/imunologia , Células Eucarióticas/metabolismo , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Legionella/classificação , Legionella/genética , Legionella/imunologia , Legionella pneumophila/genética , Legionella pneumophila/imunologia , Doença dos Legionários/imunologia , Doença dos Legionários/patologia
3.
Curr Opin Immunol ; 60: 71-80, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31176142

RESUMO

Inside host cells, guanylate binding proteins (GBPs) rapidly assemble into large antimicrobial defense complexes that combat a wide variety of bacterial pathogens. These massive nanomachines often completely coat targeted microbes where they act as recruitment platforms for downstream effectors capable of direct bactericidal activity. GBP-containing platforms also serve as sensory hubs to activate inflammasome-driven responses in the mammalian cytosol while in plants like Arabidopsis, GBP orthologues may facilitate intranuclear signaling for immunity against invasive phytopathogens. Together, this group of immune GTPases serve as a major defensive repertoire to protect the host cell interior from bacterial colonization across plant and animal kingdoms.


Assuntos
Células Eucarióticas/imunologia , Células Eucarióticas/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Imunidade , Interferons/metabolismo , Animais , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Infecções Bacterianas/metabolismo , Infecções Bacterianas/microbiologia , Evolução Biológica , Células Eucarióticas/microbiologia , Proteínas de Ligação ao GTP/química , Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Humanos , Doenças das Plantas , Fenômenos Fisiológicos Vegetais , Plantas/genética , Plantas/imunologia , Plantas/metabolismo
4.
Artigo em Inglês | MEDLINE | ID: mdl-31058096

RESUMO

Over the last decade, there has been significant advances in the understanding of the cross-talk between metabolism and immune responses. It is now evident that immune cell effector function strongly depends on the metabolic pathway in which cells are engaged in at a particular point in time, the activation conditions, and the cell microenvironment. It is also clear that some metabolic intermediates have signaling as well as effector properties and, hence, topics such as immunometabolism, metabolic reprograming, and metabolic symbiosis (among others) have emerged. Viruses completely rely on their host's cell energy and molecular machinery to enter, multiply, and exit for a new round of infection. This review explores how viruses mimic, exploit or interfere with host cell metabolic pathways and how, in doing so, they may evade immune responses. It offers a brief outline of key metabolic pathways, mitochondrial function and metabolism-related signaling pathways, followed by examples of the mechanisms by which several viral proteins regulate host cell metabolic activity.


Assuntos
Células Eucarióticas/virologia , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Imunidade Celular , Proteínas Virais/metabolismo , Fatores de Virulência/metabolismo , Vírus/crescimento & desenvolvimento , Células Eucarióticas/imunologia , Células Eucarióticas/metabolismo , Metabolismo , Vírus/imunologia , Vírus/patogenicidade
5.
Wiley Interdiscip Rev RNA ; 10(4): e1535, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30963709

RESUMO

Small RNAs and their associated RNA interference (RNAi) pathways underpin diverse mechanisms of gene regulation and genome defense across all three kingdoms of life and are integral to virus-host interactions. In plants, fungi and many animals, an ancestral RNAi pathway exists as a host defense mechanism whereby viral double-stranded RNA is processed to small RNAs that enable recognition and degradation of the virus. While this antiviral RNAi pathway is not generally thought to be present in mammals, other RNAi mechanisms can influence infection through both viral- and host-derived small RNAs. Furthermore, a burgeoning body of data suggests that small RNAs in mammals can function in a non-cell autonomous manner to play various roles in cell-to-cell communication and disease through their transport in extracellular vesicles. While vesicular small RNAs have not been proposed as an antiviral defense pathway per se, there is increasing evidence that the export of host- or viral-derived RNAs from infected cells can influence various aspects of the infection process. This review discusses the current knowledge of extracellular RNA functions in viral infection and the technical challenges surrounding this field of research. This article is categorized under: Regulatory RNAs/RNAi/Riboswitches > Regulatory RNAs RNA in Disease and Development > RNA in Disease Regulatory RNAs/RNAi/Riboswitches > RNAi: Mechanisms of Action.


Assuntos
Células Eucarióticas/imunologia , Células Eucarióticas/virologia , Vesículas Extracelulares/metabolismo , Regulação da Expressão Gênica , Interações entre Hospedeiro e Microrganismos , Interferência de RNA , Pequeno RNA não Traduzido/metabolismo
6.
RNA ; 25(7): 869-880, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31019095

RESUMO

Bacterial RNA has emerged as an important activator of innate immune responses by stimulating Toll-like receptors TLR7 and TLR8 in humans. Guanosine 2'-O-methylation at position 18 (Gm18) in bacterial tRNA was shown to antagonize tRNA-induced TLR7/8 activation, suggesting a potential role of Gm18 as an immune escape mechanism. This modification also occurs in eukaryotic tRNA, yet a physiological immune function remained to be tested. We therefore set out to investigate the immune modulatory role of Gm18 in both prokaryotic and eukaryotic microorganisms, Escherichia coli and Saccharomyces cerevisiae, and in human cells. Using RiboMethSeq analysis we show that mutation of trmH in E. coli, trm3 in S. cereviase, and CRISPR/Cas9-induced knockout of TARBP1 in H. sapiens results in loss of Gm18 within tRNA. Lack of Gm18 across the kingdoms resulted in increased immunostimulation of peripheral blood mononuclear cells when activated by tRNA preparations. In E. coli, lack of 2'-O-methyltransferase trmH also enhanced immune stimulatory properties by whole cellular RNA. In contrast, lack of Gm18 in yeasts and human cells did not affect immunostimulation by whole RNA preparations. When using live E. coli bacteria, lack of trmH did not affect overall immune stimulation although we detected a defined TLR8/RNA-dependent gene expression signature upon E. coli infection. Together, these results demonstrate that Gm18 is a global immune inhibitory tRNA modification across the kingdoms and contributes to tRNA recognition by innate immune cells, but as an individual modification has insufficient potency to modulate recognition of the investigated microorganisms.


Assuntos
Endossomos/metabolismo , Células Eucarióticas/imunologia , Guanosina/química , Imunidade Inata/imunologia , Células Procarióticas/imunologia , RNA de Transferência/metabolismo , Receptores Toll-Like/metabolismo , Células Eucarióticas/metabolismo , Humanos , Metilação , Células Procarióticas/metabolismo , RNA de Transferência/genética , Receptores Toll-Like/genética , tRNA Metiltransferases/genética , tRNA Metiltransferases/metabolismo
7.
Nat Rev Immunol ; 19(1): 31-44, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30301972

RESUMO

Protection against microbial infection in eukaryotes is provided by diverse cellular and molecular mechanisms. Here, we present a comparative view of the antiviral activity of virus-derived small interfering RNAs in fungi, plants, invertebrates and mammals, detailing the mechanisms for their production, amplification and activity. We also highlight the recent discovery of viral PIWI-interacting RNAs in animals and a new role for mobile host and pathogen small RNAs in plant defence against eukaryotic pathogens. In turn, viruses that infect plants, insects and mammals, as well as eukaryotic pathogens of plants, have evolved specific virulence proteins that suppress RNA interference (RNAi). Together, these advances suggest that an antimicrobial function of the RNAi pathway is conserved across eukaryotic kingdoms.


Assuntos
Anti-Infecciosos/imunologia , RNA Interferente Pequeno/imunologia , Animais , Células Eucarióticas/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Mamíferos/imunologia , Plantas/imunologia , Interferência de RNA/imunologia
8.
J Virol ; 92(19)2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30021896

RESUMO

Due to their roles in the regulation of programmed cell death and inflammation, the cellular caspase proteases are considered antiviral factors. However, recent studies have revealed examples of proviral functions for caspases. Here, we review a growing body of literature on the role of caspases in promoting the replication of human gammaherpesviruses. We propose that gammaherpesviruses have evolved ways to redirect these enzymes and to use their activation to support viral replication and immune evasion.


Assuntos
Caspases/genética , Células Eucarióticas/virologia , Gammaherpesvirinae/genética , Proteínas Imediatamente Precoces/genética , Evasão da Resposta Imune/genética , Provírus/genética , Animais , Apoptose , Caspases/imunologia , Células Eucarióticas/imunologia , Células Eucarióticas/metabolismo , Evolução Molecular , Gammaherpesvirinae/imunologia , Gammaherpesvirinae/metabolismo , Regulação da Expressão Gênica , Humanos , Proteínas Imediatamente Precoces/imunologia , Provírus/imunologia , Provírus/metabolismo , Transdução de Sinais , Vírion/genética , Vírion/imunologia , Vírion/metabolismo , Replicação Viral
9.
Curr Issues Mol Biol ; 25: 43-60, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28875939

RESUMO

Following colonization of host tissues, bacterial pathogens encounter new niches in which they must gain access to nutrients and cope with stresses and defence signals generated by the host. For some pathogens, the adaptation to a new 'within-host' lifestyle involves modifications of envelope components that bear molecular patterns normally recognized by the host innate immune system. These new modified patterns limit host recognition, therefore promoting immune evasion and pathogenicity. In this review, we describe how envelope components like the peptidoglycan or lipopolysaccharide can be altered within the host to impair responses triggered by pattern recognition receptors (PRR). We also discuss the few cases reported to date of chemical modifications that occur in the envelope of some intracellular bacterial pathogens when they reside inside eukaryotic cells. These envelope alterations may have evolved due to the sentinel role performed by PRRs over pathogen-specific molecular patterns. The available data indicate that only selected pathogens seem to evade recognition due to 'within-host' envelope changes, with most of them displaying such patterns also in non host environments. Given the importance of these alterations, future studies should focus in the responsible pathogen regulators, most yet unknown, that could be targeted to prevent immune evasion.


Assuntos
Cápsulas Bacterianas/química , Lipopolissacarídeos/imunologia , Antígeno 96 de Linfócito/imunologia , Proteínas NLR/imunologia , Peptidoglicano/imunologia , Receptores Toll-Like/imunologia , Animais , Bactérias/crescimento & desenvolvimento , Bactérias/imunologia , Cápsulas Bacterianas/imunologia , Células Eucarióticas/imunologia , Células Eucarióticas/microbiologia , Regulação da Expressão Gênica , Humanos , Evasão da Resposta Imune , Imunidade Inata , Inflamassomos/imunologia , Inflamassomos/metabolismo , Lipopolissacarídeos/metabolismo , Antígeno 96 de Linfócito/genética , Proteínas NLR/genética , Peptidoglicano/metabolismo , Transdução de Sinais , Receptores Toll-Like/genética
10.
Artigo em Inglês | MEDLINE | ID: mdl-28432128

RESUMO

In contrast to live attenuated vaccines, which are designed to induce immunity through a time-limited bloom in systemic tissues, the microbiota is a persistent feature of body surfaces, especially the intestine. The immune responses to the microbiota are idiosyncratic depending on the niche intimacy of different taxa and generally adapt the host to avoid overgrowth and maintain mutualism rather than to eliminate the organisms of that taxon. Both the microbiota and the host have so much molecular cross talk controlling each other, that the prokaryotic and the eukaryotic spaces of the host-microbial superorganism are federal rather than sovereign. This molecular cross talk is vital for the immune system to develop its mature form. Nevertheless, the microbiota/host biomass spaces are rather well separated: The microbiota also limits colonization and penetration of pathogens through intense metabolic competition. Immune responses to those members of the microbiota mutually adapted to intimate association at mucosal surfaces have attractive potential durability, but for clinical use as persistent vehicles they would require personalization and engineered reversibility to manage the immune context and complications in individual human subjects.


Assuntos
Interações Hospedeiro-Patógeno , Infecções/imunologia , Microbiota/imunologia , Vacinas/imunologia , Animais , Células Eucarióticas/imunologia , Humanos , Células Procarióticas/imunologia
11.
J Cell Biol ; 216(12): 3931-3948, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29097627

RESUMO

Intracellular bacterial pathogens have developed versatile strategies to generate niches inside the eukaryotic cells that allow them to survive and proliferate. Making a home inside the host offers many advantages; however, intracellular bacteria must also overcome many challenges, such as disarming innate immune signaling and accessing host nutrient supplies. Gaining entry into the cell and avoiding degradation is only the beginning of a successful intracellular lifestyle. To establish these replicative niches, intracellular pathogens secrete various virulence proteins, called effectors, to manipulate host cell signaling pathways and subvert host defense mechanisms. Many effectors mimic host enzymes, whereas others perform entirely novel enzymatic functions. A large volume of work has been done to understand how intracellular bacteria manipulate membrane trafficking pathways. In this review, we focus on how intracellular bacterial pathogens target innate immune signaling, the unfolded protein response, autophagy, and cellular metabolism and exploit these pathways to their advantage. We also discuss how bacterial pathogens can alter host gene expression by directly modifying histones or hijacking the ubiquitination machinery to take control of several host signaling pathways.


Assuntos
Bactérias/metabolismo , Células Eucarióticas/microbiologia , Interações Hospedeiro-Patógeno , Resposta a Proteínas não Dobradas/imunologia , Autofagia/genética , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Transporte Biológico , Células Eucarióticas/imunologia , Células Eucarióticas/metabolismo , Regulação da Expressão Gênica , Histonas/genética , Histonas/imunologia , Humanos , Imunidade Inata , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/imunologia , Transdução de Sinais , Ubiquitinação
12.
Biol Direct ; 12(1): 5, 2017 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-28187792

RESUMO

Complementarity between nucleic acid molecules is central to biological information transfer processes. Apart from the basal processes of replication, transcription and translation, complementarity is also employed by multiple defense and regulatory systems. All cellular life forms possess defense systems against viruses and mobile genetic elements, and in most of them some of the defense mechanisms involve small guide RNAs or DNAs that recognize parasite genomes and trigger their inactivation. The nucleic acid-guided defense systems include prokaryotic Argonaute (pAgo)-centered innate immunity and CRISPR-Cas adaptive immunity as well as diverse branches of RNA interference (RNAi) in eukaryotes. The archaeal pAgo machinery is the direct ancestor of eukaryotic RNAi that, however, acquired additional components, such as Dicer, and enormously diversified through multiple duplications. In contrast, eukaryotes lack any heritage of the CRISPR-Cas systems, conceivably, due to the cellular toxicity of some Cas proteins that would get activated as a result of operon disruption in eukaryotes. The adaptive immunity function in eukaryotes is taken over partly by the PIWI RNA branch of RNAi and partly by protein-based immunity. In this review, I briefly discuss the interplay between homology and analogy in the evolution of RNA- and DNA-guided immunity, and attempt to formulate some general evolutionary principles for this ancient class of defense systems. REVIEWERS: This article was reviewed by Mikhail Gelfand and Bojan Zagrovic.


Assuntos
Imunidade Adaptativa/genética , Células Eucarióticas/imunologia , Evolução Molecular , Imunidade Inata/genética , Células Procarióticas/imunologia , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Proteínas Argonautas/fisiologia , Sistemas CRISPR-Cas/fisiologia , DNA/imunologia , RNA/imunologia , Interferência de RNA
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(8): 1026-30, 2016 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-27412930

RESUMO

Objective To design, construct and express a chimeric αß TCR harboring the immunoglobulin-like (Ig) domain of γδ TCR in Jurkat T cells. Methods The fusion sites of TCR δIg were determined by bioinformatics analysis. Then the protein structures of TCR α δIg and TCR ß Î´Ig were predicted by homology modeling. Furthermore, the structures of TCR α δIg and TCR ß Î´Ig were compared with the wild type (wt) TCR α and TCR ß respectively by combinatorial extension (CE). After that, the TCR α δIg and TCR ß Î´Ig were fused to fluorescent protein ECFP and EYFP respectively via the overlap PCR, and then the fusion genes (TCR α δIg-ECFP and TCR ß Î´Ig-EYFP) were cloned into pIRES2-EGFP vector and respectively located at the upstream and downstream of an internal ribosome entry site (IRES). The recombinant prokaryotic expression vector pIRES-TCR ßδIg-EYFP/TCR αδIg-ECFP was transferred into Jurkat T cells. Finally, the expression of TCR δIg in Jurkat T cells was monitored by confocal laser scanning microscopy (CLSM). Results The variable region structure of the TCR δIg did not change and the antigen recognition active regions remained stable compared to the wtTCR. The recombinant expression plasmid was successfully constructed as confirmed by PCR identification and sequencing analysis. CLSM showed that TCR δIg was expressed and located at the plasma membrane of Jurkat T cells. Conclusion The design of TCR δIg was reasonable and the TCR δIg could be expressed on Jurkat T cell surface.


Assuntos
Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/imunologia , Sequência de Aminoácidos , Células Eucarióticas/imunologia , Células Eucarióticas/metabolismo , Expressão Gênica/imunologia , Humanos , Células Jurkat , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microscopia Confocal , Modelos Moleculares , Domínios Proteicos , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/genética , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Linfócitos T/metabolismo
14.
Med Sci (Paris) ; 32(6-7): 640-5, 2016.
Artigo em Francês | MEDLINE | ID: mdl-27406776

RESUMO

Since genetics has shown that mutation predates selection, biology has developed within the Darwinian paradigm framework. However, a mechanism that produces favorable mutations preferentially in response to adaptive constraints has been recently identified. This mechanism, the CRISPR-Cas adaptive immunity system, is considered as a bona fide example of Lamarckian evolution, even if it only reflects loosely Lamarck's ideas. This unusual evolutionary process is made possible by two prokaryotic properties: i) somatic and germinal cells are not distinct sets of cells; ii) Archae and Bacteria very frequently integrate DNA fragments from the environment, and they therefore have access to a source of "ready-made" useful genetic information. The CRISPR-Cas is a defense system against viruses and plasmids that is based on the integration of genomic fragments of these infectious agents into the host genome, and that protects the host against subsequent infections. Therefore, this mechanism does produce advantageous mutations by integrating DNA from the environment and allowing its transmission to descendants. In conclusion, most of the time evolution relies on purely Darwinian processes, i.e. mutations occurring at random, but in a small minority of cases the occurrence of mutations is more or less biased, and is therefore more or less Lamarckian. Although they are rare, such processes are nevertheless important to our understanding of the plurality of modes of evolution.


Assuntos
Imunidade Adaptativa/genética , Sistemas CRISPR-Cas/fisiologia , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/fisiologia , Evolução Molecular , Técnicas Genéticas , Mutagênese Sítio-Dirigida , Animais , Células Eucarióticas/imunologia , Células Eucarióticas/metabolismo , Técnicas Genéticas/tendências , Humanos , Mutagênese Sítio-Dirigida/métodos , Mutagênese Sítio-Dirigida/tendências , Células Procarióticas/imunologia , Células Procarióticas/metabolismo
15.
Trends Microbiol ; 24(4): 294-306, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26852268

RESUMO

Clustered, regularly interspaced, short palindromic repeats - CRISPR-associated (CRISPR-Cas) systems - are sequence-specific RNA-directed endonuclease complexes that bind and cleave nucleic acids. These systems evolved within prokaryotes as adaptive immune defenses to target and degrade nucleic acids derived from bacteriophages and other foreign genetic elements. The antiviral function of these systems has now been exploited to combat eukaryotic viruses throughout the viral life cycle. Here we discuss current advances in CRISPR-Cas9 technology as a eukaryotic antiviral defense.


Assuntos
Células Eucarióticas/imunologia , Células Procarióticas/imunologia , Vírus/imunologia , Imunidade Adaptativa/imunologia , Animais , Bacteriófagos/imunologia , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/imunologia , Geminiviridae/genética , Geminiviridae/imunologia , HIV/genética , HIV/imunologia , Vírus da Hepatite B/genética , Vírus da Hepatite B/imunologia , Humanos , Sistema Imunitário , Modelos Moleculares , Papillomaviridae/genética , Papillomaviridae/imunologia , Vírus/genética
16.
Nihon Saikingaku Zasshi ; 69(3): 503-11, 2014.
Artigo em Japonês | MEDLINE | ID: mdl-25186641

RESUMO

Legionella are gram-negative bacteria ubiquitously found in freshwater and soil environments. Once inhaled by humans, Legionella infection could result in a severe form of pneumonia known as Legionellosis. Legionella translocate ~300 effector proteins into host cells via the Dot/Icm type IV secretion system, which is central to Legionella pathogenesis. Here I describe a brief review on recent advances in research on the molecular basis of Legionella-eukaryotic-cell interaction.


Assuntos
Proteínas de Bactérias/imunologia , Proteínas de Bactérias/fisiologia , Sistemas de Secreção Bacterianos/imunologia , Sistemas de Secreção Bacterianos/fisiologia , Células Eucarióticas/imunologia , Células Eucarióticas/microbiologia , Interações Hospedeiro-Patógeno/imunologia , Legionella pneumophila/genética , Legionella pneumophila/imunologia , Doença dos Legionários/imunologia , Doença dos Legionários/microbiologia , Animais , Água Doce/microbiologia , Humanos , Legionella pneumophila/patogenicidade , Macrófagos Alveolares/microbiologia , Filogenia , Microbiologia do Solo
17.
Brief Bioinform ; 14(6): 753-74, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23097412

RESUMO

In this article, a framework for an in silico pipeline is presented as a guide to high-throughput vaccine candidate discovery for eukaryotic pathogens, such as helminths and protozoa. Eukaryotic pathogens are mostly parasitic and cause some of the most damaging and difficult to treat diseases in humans and livestock. Consequently, these parasitic pathogens have a significant impact on economy and human health. The pipeline is based on the principle of reverse vaccinology and is constructed from freely available bioinformatics programs. There are several successful applications of reverse vaccinology to the discovery of subunit vaccines against prokaryotic pathogens but not yet against eukaryotic pathogens. The overriding aim of the pipeline, which focuses on eukaryotic pathogens, is to generate through computational processes of elimination and evidence gathering a ranked list of proteins based on a scoring system. These proteins are either surface components of the target pathogen or are secreted by the pathogen and are of a type known to be antigenic. No perfect predictive method is yet available; therefore, the highest-scoring proteins from the list require laboratory validation.


Assuntos
Células Eucarióticas/imunologia , Vacinas , Simulação por Computador
18.
J Immunol ; 187(9): 4403-9, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22013206

RESUMO

The unfolded protein response (UPR) is a eukaryotic cellular adaptive mechanism that functions to cope with stress of the endoplasmic reticulum (ER). Accumulating evidence demonstrates that the tumor microenvironment contains stressors that elicit a UPR, which has been demonstrated to be a cell-intrinsic mechanism crucial for tumorigenesis. In addition, the UPR is a source of proinflammatory signaling whose downstream mediators may hamper antitumor immunity. We discuss how the UPR may impair Ag presentation, which could result in defective T cell priming, also leading to tumor escape and growth. Further, we discuss the recent finding that ER stress and attendant proinflammation can be transmitted from ER-stressed tumor cells to myeloid cells. The ideas presented suggest that, in addition to being a cell-intrinsic mechanism of tumor survival, the tumor UPR can serve as a cell-extrinsic regulator of tumorigenesis by remodeling the immune response in the tumor microenvironment.


Assuntos
Retículo Endoplasmático/imunologia , Retículo Endoplasmático/patologia , Células Eucarióticas/imunologia , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Estresse Oxidativo/imunologia , Microambiente Tumoral/imunologia , Resposta a Proteínas não Dobradas/imunologia , Animais , Linhagem Celular Tumoral , Retículo Endoplasmático/genética , Células Eucarióticas/metabolismo , Células Eucarióticas/patologia , Humanos , Neoplasias Experimentais/genética , Estresse Oxidativo/genética , Microambiente Tumoral/genética , Resposta a Proteínas não Dobradas/genética
19.
Biol Chem ; 389(5): 579-83, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18953725

RESUMO

Experimental preparations based on a DNA vaccine encoding the ESAT-6 antigen of Mycobacterium tuberculosis have been obtained (KpONE6) and studied for immunogenic effects in the murine model. The core of the preparation contains DNA of the recombinant plasmid pONE6 encapsulated within a spermidine-polyglucin conjugate, thereby protecting the DNA vaccine from degradation. KpONE6 induces a proliferative T-cell immune response in mice upon intramuscular immunization.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Vacinas contra a Tuberculose/genética , Vacinas contra a Tuberculose/imunologia , Animais , Proliferação de Células , Citocinas/biossíntese , Composição de Medicamentos , Ensaio de Imunoadsorção Enzimática , Células Eucarióticas/imunologia , Injeções Intramusculares , Interferon gama/biossíntese , Interferon gama/genética , Linfócitos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ensaios de Proteção de Nucleases , Plasmídeos/genética , Plasmídeos/imunologia , Polissacarídeos/imunologia , Espermidina/imunologia , Vacinas contra a Tuberculose/administração & dosagem , Vacinas de DNA/genética , Vacinas de DNA/imunologia
20.
Chem Soc Rev ; 37(7): 1337-46, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18568160

RESUMO

Quorum sensing (QS) has traditionally referred to a mechanism of communication within a species of bacteria. However, emerging research implicates QS in interspecies communication and competition, and such systems have been proposed in a wide variety of bacteria. This activity of bacterial QS also extends to relationships between bacteria and eukaryotes and host-pathogen interactions in both clinical and agricultural settings are of particular interest. These relationships are particularly pertinent in light of the rising prevalence of antibiotic resistant bacteria. In this tutorial review we describe bacterial QS and its capacity in interspecies and interkingdom interactions, as well as the corresponding eukaryotic responses.


Assuntos
Antibacterianos/farmacologia , Bactérias/metabolismo , Comunicação Celular/fisiologia , Células Eucarióticas/fisiologia , Percepção de Quorum/fisiologia , Antibacterianos/química , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Comunicação Celular/genética , Células Eucarióticas/imunologia , Regulação Bacteriana da Expressão Gênica , Percepção de Quorum/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...