Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Bacteriol ; : e0009124, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38842342

RESUMO

Pathogenic species within the Rickettsia genus are transmitted to humans through arthropod vectors and cause a spectrum of diseases ranging from mild to life-threatening. Despite rickettsiae posing an emerging global health risk, the genetic requirements of their infectious life cycles remain poorly understood. A major hurdle toward building this understanding has been the lack of efficient tools for genetic manipulation, owing to the technical difficulties associated with their obligate intracellular nature. To this end, we implemented the Tet-On system to enable conditional gene expression in Rickettsia parkeri. Using Tet-On, we show inducible expression of antibiotic resistance and a fluorescent reporter. We further used this inducible promoter to screen the ability of R. parkeri to express four variants of the catalytically dead Cas9 (dCas9). We demonstrate that all four dCas9 variants can be expressed in R. parkeri and used for CRISPR interference (CRISPRi)-mediated targeted gene knockdown. We show targeted knockdown of an antibiotic resistance gene as well as the endogenous virulence factor sca2. Altogether, we have developed systems for inducible gene expression and CRISPRi-mediated gene knockdown for the first time in rickettsiae, laying the groundwork for more scalable, targeted mechanistic investigations into their infectious life cycles.IMPORTANCEThe spotted fever group of Rickettsia contains vector-borne pathogenic bacteria that are neglected and emerging threats to public health. Due to the obligate intracellular nature of rickettsiae, the development of tools for genetic manipulation has been stunted, and the molecular and genetic underpinnings of their infectious lifecycle remain poorly understood. Here, we expand the genetic toolkit by introducing systems for conditional gene expression and CRISPR interference (CRISPRi)-mediated gene knockdown. These systems allow for relatively easy manipulation of rickettsial gene expression. We demonstrate the effectiveness of these tools by disrupting the intracellular life cycle using CRISPRi to deplete the sca2 virulence factor. These tools will be crucial for building a more comprehensive and detailed understanding of rickettsial biology and pathogenesis.

2.
bioRxiv ; 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38559073

RESUMO

Pathogenic species within the Rickettsia genus are transmitted to humans through arthropod vectors and cause a spectrum of diseases ranging from mild to life-threatening. Despite rickettsiae posing an emerging global health risk, the genetic requirements of their infectious life cycles remain poorly understood. A major hurdle toward building this understanding has been the lack of efficient tools for genetic manipulation, owing to the technical difficulties associated with their obligate intracellular nature. To this end, we implemented the Tet-On system to enable conditional gene expression in Rickettsia parkeri. Using Tet-On, we show inducible expression of antibiotic resistance and a fluorescent reporter. We further used this inducible promoter to screen the ability of R. parkeri to express four variants of the catalytically dead Cas9 (dCas9). We demonstrate that all four dCas9 variants can be expressed in R. parkeri and used for CRISPR interference (CRISPRi)-mediated targeted gene knockdown. We show targeted knockdown of an antibiotic resistance gene as well as the endogenous virulence factor sca2. Altogether, we have developed systems for inducible gene expression and CRISPRi-mediated gene knockdown for the first time in rickettsiae, laying the groundwork for more scalable, targeted mechanistic investigations into their infectious life cycles.

3.
J Bacteriol ; 206(2): e0040423, 2024 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-38315013

RESUMO

Our understanding of free-living bacterial models like Escherichia coli far outpaces that of obligate intracellular bacteria, which cannot be cultured axenically. All obligate intracellular bacteria are host-associated, and many cause serious human diseases. Their constant exposure to the distinct biochemical niche of the host has driven the evolution of numerous specialized bacteriological and genetic adaptations, as well as innovative molecular mechanisms of infection. Here, we review the history and use of pathogenic Rickettsia species, which cause an array of vector-borne vascular illnesses, as model systems to probe microbial biology. Although many challenges remain in our studies of these organisms, the rich pathogenic and biological diversity of Rickettsia spp. constitutes a unique backdrop to investigate how microbes survive and thrive in host and vector cells. We take a bacterial-focused perspective and highlight emerging insights that relate to new host-pathogen interactions, bacterial physiology, and evolution. The transformation of Rickettsia spp. from pathogens to models demonstrates how recalcitrant microbes may be leveraged in the lab to tap unmined bacterial diversity for new discoveries. Rickettsia spp. hold great promise as model systems not only to understand other obligate intracellular pathogens but also to discover new biology across and beyond bacteria.


Assuntos
Rickettsia , Humanos , Rickettsia/genética , Interações Hospedeiro-Patógeno , Biologia
4.
bioRxiv ; 2023 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-38014272

RESUMO

Pathogenic bacteria secrete protein effectors to hijack host machinery and remodel their infectious niche. Rickettsia spp. are obligate intracellular bacteria that can cause life-threatening disease, but their absolute dependence on the host cell environment has impeded discovery of rickettsial effectors and their host targets. We implemented bioorthogonal non-canonical amino acid tagging (BONCAT) during R. parkeri infection to selectively label, isolate, and identify secreted effectors. As the first use of BONCAT in an obligate intracellular bacterium, our screen more than doubles the number of experimentally validated effectors for R. parkeri. The novel secreted rickettsial factors (Srfs) we identified include Rickettsia-specific proteins of unknown function that localize to the host cytoplasm, mitochondria, and ER. We further show that one such effector, SrfD, interacts with the host Sec61 translocon. Altogether, our work uncovers a diverse set of previously uncharacterized rickettsial effectors and lays the foundation for a deeper exploration of the host-pathogen interface.

5.
bioRxiv ; 2023 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-37333133

RESUMO

Interorganelle communication regulates cellular homeostasis through the formation of tightly-associated membrane contact sites 1-3. Prior work has identified several ways that intracellular pathogens alter contacts between eukaryotic membranes 4-6, but there is no existing evidence for contact sites spanning eukaryotic and prokaryotic membranes. Here, using a combination of live-cell microscopy and transmission and focused-ion-beam scanning electron microscopy, we demonstrate that the intracellular bacterial pathogen Rickettsia parkeri forms a direct membrane contact site between its bacterial outer membrane and the rough endoplasmic reticulum (ER), with tethers that are approximately 55 nm apart. Depletion of the ER-specific tethers VAPA and VAPB reduced the frequency of rickettsia-ER contacts, suggesting these interactions mimic organelle-ER contacts. Overall, our findings illuminate a direct, interkingdom membrane contact site uniquely mediated by rickettsia that seems to mimic traditional host MCSs.

6.
J Bacteriol ; 204(7): e0018222, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35727033

RESUMO

Rickettsia spp. are obligate intracellular bacterial pathogens that have evolved a variety of strategies to exploit their host cell niche. However, the bacterial factors that contribute to this intracellular lifestyle are poorly understood. Here, we show that the conserved ankyrin repeat protein RARP-1 supports Rickettsia parkeri infection. Specifically, RARP-1 promotes efficient host cell entry and growth within the host cytoplasm, but it is not necessary for cell-to-cell spread or evasion of host autophagy. We further demonstrate that RARP-1 is not secreted into the host cytoplasm by R. parkeri. Instead, RARP-1 resides in the periplasm, and we identify several binding partners that are predicted to work in concert with RARP-1 during infection. Altogether, our data reveal that RARP-1 plays a critical role in the rickettsial life cycle. IMPORTANCERickettsia spp. are obligate intracellular bacterial pathogens that pose a growing threat to human health. Nevertheless, their strict reliance on a host cell niche has hindered investigation of the molecular mechanisms driving rickettsial infection. This study yields much-needed insight into the Rickettsia ankyrin repeat protein RARP-1, which is conserved across the genus but has not yet been functionally characterized. Earlier work had suggested that RARP-1 is secreted into the host cytoplasm. However, the results from this work demonstrate that R. parkeri RARP-1 resides in the periplasm and is important both for invasion of host cells and for growth in the host cell cytoplasm. These results reveal RARP-1 as a novel regulator of the rickettsial life cycle.


Assuntos
Periplasma , Rickettsia , Repetição de Anquirina , Citoplasma , Humanos , Rickettsia/genética , Rickettsia/metabolismo
7.
Trends Microbiol ; 30(5): 511-512, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35115187
8.
Pathog Dis ; 79(4)2021 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-33784388

RESUMO

Rickettsiae are obligate intracellular bacteria that can cause life-threatening illnesses and are among the oldest known vector-borne pathogens. Members of this genus are extraordinarily diverse and exhibit a broad host range. To establish intracellular infection, Rickettsia species undergo complex, multistep life cycles that are encoded by heavily streamlined genomes. As a result of reductive genome evolution, rickettsiae are exquisitely tailored to their host cell environment but cannot survive extracellularly. This host-cell dependence makes for a compelling system to uncover novel host-pathogen biology, but it has also hindered experimental progress. Consequently, the molecular details of rickettsial biology and pathogenesis remain poorly understood. With recent advances in molecular biology and genetics, the field is poised to start unraveling the molecular mechanisms of these host-pathogen interactions. Here, we review recent discoveries that have shed light on key aspects of rickettsial biology. These studies have revealed that rickettsiae subvert host cells using mechanisms that are distinct from other better-studied pathogens, underscoring the great potential of the Rickettsia genus for revealing novel biology. We also highlight several open questions as promising areas for future study and discuss the path toward solving the fundamental mysteries of this neglected and emerging human pathogen.


Assuntos
Proteínas de Bactérias/genética , Genoma Bacteriano , Especificidade de Hospedeiro/genética , Estágios do Ciclo de Vida/genética , Infecções por Rickettsia/microbiologia , Rickettsia/genética , Animais , Proteínas de Bactérias/classificação , Proteínas de Bactérias/metabolismo , Elementos de DNA Transponíveis , Regulação Bacteriana da Expressão Gênica , Humanos , Doenças Negligenciadas/microbiologia , Doenças Negligenciadas/patologia , Mapeamento de Interação de Proteínas , Rickettsia/crescimento & desenvolvimento , Rickettsia/metabolismo , Rickettsia/patogenicidade , Infecções por Rickettsia/patologia , Sistemas de Secreção Tipo IV/genética , Sistemas de Secreção Tipo IV/metabolismo
9.
Mol Biol Cell ; 30(17): 2124-2133, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31242077

RESUMO

Listeria monocytogenes is a human bacterial pathogen that disseminates through host tissues using a process called cell-to-cell spread. This critical yet understudied virulence strategy resembles a vesicular form of intercellular trafficking that allows L. monocytogenes to move between host cells without escaping the cell. Interestingly, eukaryotic cells can also directly exchange cellular components via intercellular communication pathways (e.g., trans-endocytosis) using cell-cell adhesion, membrane trafficking, and membrane remodeling proteins. Therefore, we hypothesized that L. monocytogenes would hijack these types of host proteins during spread. Using a focused RNA interference screen, we identified 22 host genes that are important for L. monocytogenes spread. We then found that caveolins (CAV1 and CAV2) and the membrane sculpting F-BAR protein PACSIN2 promote L. monocytogenes protrusion engulfment during spread, and that PACSIN2 specifically localizes to protrusions. Overall, our study demonstrates that host intercellular communication pathways may be coopted during bacterial spread and that specific trafficking and membrane remodeling proteins promote bacterial protrusion resolution.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Caveolinas/metabolismo , Listeria monocytogenes/patogenicidade , Listeriose/genética , Listeriose/microbiologia , Células A549 , Actinas/metabolismo , Proteínas de Bactérias/metabolismo , Caveolina 1/metabolismo , Caveolina 2/metabolismo , Endocitose/fisiologia , Células Eucarióticas/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Listeria monocytogenes/metabolismo , Listeriose/metabolismo , Proteínas de Membrana/metabolismo , Transporte Proteico , Interferência de RNA , Virulência
10.
PLoS One ; 13(5): e0197012, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29723287

RESUMO

The rickettsiae are obligate intracellular alphaproteobacteria that exhibit a complex infectious life cycle in both arthropod and mammalian hosts. As obligate intracellular bacteria, rickettsiae are highly adapted to living inside a variety of host cells, including vascular endothelial cells during mammalian infection. Although it is assumed that the rickettsiae produce numerous virulence factors that usurp or disrupt various host cell pathways, they have been challenging to genetically manipulate to identify the key bacterial factors that contribute to infection. Motivated to overcome this challenge, we sought to expand the repertoire of available rickettsial loss-of-function mutants, using an improved mariner-based transposon mutagenesis scheme. Here, we present the isolation of over 100 transposon mutants in the spotted fever group species Rickettsia parkeri. Transposon insertions disrupted genes whose products are implicated in a variety of pathways, including bacterial replication and metabolism, the type IV secretion system, factors with previously established roles in host cell interactions and pathogenesis, or are of unknown function. Given the need to identify critical virulence factors, forward genetic screens such as this will provide an excellent platform to more directly investigate rickettsial biology and pathogenesis.


Assuntos
Proteínas de Bactérias/genética , Elementos de DNA Transponíveis , Rickettsia/genética , Sistemas de Secreção Tipo IV/genética , Fatores de Virulência/genética , Animais , Proteínas de Bactérias/metabolismo , Chlorocebus aethiops , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Mutagênese , Mutação , Plasmídeos/química , Plasmídeos/metabolismo , Reação em Cadeia da Polimerase , Rickettsia/metabolismo , Rickettsia/patogenicidade , Transposases/genética , Transposases/metabolismo , Sistemas de Secreção Tipo IV/metabolismo , Células Vero , Fatores de Virulência/metabolismo
11.
Curr Opin Microbiol ; 35: 48-57, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27997855

RESUMO

Subversion of the host actin cytoskeleton is a critical virulence mechanism used by a variety of intracellular bacterial pathogens during their infectious life cycles. These pathogens manipulate host actin to promote actin-based motility and coordinate motility with cell-to-cell spread. Growing evidence suggests that the tactics employed by pathogens are surprisingly diverse. Here, we review recent advances suggesting that bacterial surface proteins exhibit divergent biochemical mechanisms of actin polymerization and recruit distinct host protein networks to drive motility, and that bacteria deploy secreted effector proteins that alter host cell mechanotransduction pathways to enable spread. Further investigation into the divergent strategies used by bacterial pathogens to mobilize actin will reveal new insights into pathogenesis and cytoskeleton regulation.


Assuntos
Citoesqueleto de Actina/fisiologia , Actinas/fisiologia , Bactérias/patogenicidade , Infecções Bacterianas/microbiologia , Proteínas de Bactérias/metabolismo , Animais , Bactérias/metabolismo , Fenômenos Fisiológicos Bacterianos , Citoplasma/microbiologia , Interações Hospedeiro-Patógeno , Locomoção , Camundongos , Microtúbulos , Virulência
12.
Cell ; 167(3): 670-683.e10, 2016 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-27768890

RESUMO

Spotted fever group (SFG) rickettsiae are human pathogens that infect cells in the vasculature. They disseminate through host tissues by a process of cell-to-cell spread that involves protrusion formation, engulfment, and vacuolar escape. Other bacterial pathogens rely on actin-based motility to provide a physical force for spread. Here, we show that SFG species Rickettsia parkeri typically lack actin tails during spread and instead manipulate host intercellular tension and mechanotransduction to promote spread. Using transposon mutagenesis, we identified surface cell antigen 4 (Sca4) as a secreted effector of spread that specifically promotes protrusion engulfment. Sca4 interacts with the cell-adhesion protein vinculin and blocks association with vinculin's binding partner, α-catenin. Using traction and monolayer stress microscopy, we show that Sca4 reduces vinculin-dependent mechanotransduction at cell-cell junctions. Our results suggest that Sca4 relieves intercellular tension to promote protrusion engulfment, which represents a distinctive strategy for manipulating cytoskeletal force generation to enable spread.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno , Mecanotransdução Celular , Infecções por Rickettsia/metabolismo , Infecções por Rickettsia/microbiologia , Rickettsia/patogenicidade , Vinculina/metabolismo , Actinas/metabolismo , Sequência de Aminoácidos , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Caderinas/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Elementos de DNA Transponíveis/genética , Febre/metabolismo , Febre/microbiologia , Humanos , Mutagênese Insercional , Mutação , Rickettsia/metabolismo , alfa Catenina/metabolismo
13.
Curr Biol ; 24(1): 98-103, 2014 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-24361066

RESUMO

Many intracellular bacterial pathogens undergo actin-based motility to promote cell-cell spread during infection [1]. For each pathogen, motility was assumed to be driven by a single actin polymerization pathway. Curiously, spotted fever group Rickettsia differ from other pathogens in possessing two actin-polymerizing proteins. RickA, an activator of the host Arp2/3 complex, was initially proposed to drive motility [2, 3]. Sca2, a mimic of host formins [4, 5], was later shown to be required for motility [6]. Whether and how their activities are coordinated has remained unclear. Here, we show that each protein directs an independent mode of Rickettsia parkeri motility at different times during infection. Early after invasion, motility is slow and meandering, generating short, curved actin tails that are enriched with Arp2/3 complex and cofilin. Early motility requires RickA and Arp2/3 complex and is correlated with transient RickA localization to the bacterial pole. Later in infection, motility is faster and directionally persistent, resulting in long, straight actin tails. Late motility is independent of Arp2/3 complex and RickA and requires Sca2, which accumulates at the bacterial pole. Both motility pathways facilitate cell-to-cell spread. The ability to exploit two actin assembly pathways may allow Rickettsia to establish an intracellular niche and spread between diverse cells throughout a prolonged infection.


Assuntos
Actinas/metabolismo , Rickettsia/metabolismo , Animais , Ataxinas , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Chlorocebus aethiops , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Rickettsia/citologia , Células Vero
14.
PLoS One ; 7(5): e37310, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22624012

RESUMO

Despite recent advances in our ability to genetically manipulate Rickettsia, little has been done to employ genetic tools to study the expression and localization of Rickettsia virulence proteins. Using a mariner-based Himar1 transposition system, we expressed an epitope-tagged variant of the actin polymerizing protein RickA under the control of its native promoter in Rickettsia parkeri, allowing the detection of RickA using commercially-available antibodies. Native RickA and epitope-tagged RickA exhibited similar levels of expression and were specifically localized to bacteria. To further facilitate protein expression in Rickettsia, we also developed a plasmid for Rickettsia insertion and expression (pRIE), containing a variant Himar1 transposon with enhanced flexibility for gene insertion, and used it to generate R. parkeri strains expressing diverse fluorescent proteins. Expression of epitope-tagged proteins in Rickettsia will expand our ability to assess the regulation and function of important virulence factors.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Rickettsia/metabolismo , Rickettsia/patogenicidade , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/genética , Chlorocebus aethiops , Imunofluorescência , Regulação Bacteriana da Expressão Gênica/genética , Immunoblotting , Microscopia de Fluorescência , Mapeamento Físico do Cromossomo , Plasmídeos/genética , Rickettsia/genética , Células Vero , Fatores de Virulência/genética
15.
Mol Cell ; 40(5): 798-809, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-21145487

RESUMO

T cell receptor (TCR) signaling to NF-κB is required for antigen-induced T cell activation. We conducted an expression-cloning screen for modifiers of CARD11, a critical adaptor in antigen receptor signaling, and identified the kinesin-3 family member GAKIN as a CARD11 inhibitor. GAKIN negatively regulates TCR signaling to NF-κB, associates with CARD11 in a signal-dependent manner and can compete with the required signaling protein, Bcl10, for association. In addition, GAKIN dynamically localizes to the immunological synapse and regulates the redistribution of CARD11 from the central region of the synapse to a distal region. We propose that CARD11 scaffold function and occupancy at the center of the synapse are negatively regulated by GAKIN to tune the output of antigen-receptor signaling.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/metabolismo , Guanilato Ciclase/metabolismo , Sinapses Imunológicas/metabolismo , Cinesinas/metabolismo , Transdução de Sinais/imunologia , Humanos , Células Jurkat , NF-kappa B/metabolismo , Receptores de Antígenos/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo
16.
Biochemistry ; 49(38): 8240-50, 2010 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-20799731

RESUMO

The regulated activation of NF-κB by antigen receptor signaling is required for normal B and T lymphocyte activation during the adaptive immune response. Dysregulated NF-κB activation is associated with several types of lymphoma, including diffuse large B cell lymphoma (DLBCL). During normal antigen receptor signaling, the multidomain scaffold protein CARD11 undergoes a transition from a closed, inactive state to an open, active conformation that recruits several signaling proteins into a complex, leading to IKK kinase activation. This transition is regulated by the CARD11 inhibitory domain (ID), which participates in intramolecular interactions that prevent cofactor binding to CARD11 prior to signaling, but which is neutralized after receptor engagement by phosphorylation. Several oncogenic CARD11 mutations have been identified in DLBCL that enhance activity and that are mostly found in the coiled-coil domain. However, the mechanisms by which these mutations cause CARD11 hyperactivity and spontaneous NF-κB activation are poorly understood. In this report, we provide several lines of evidence that oncogenic mutations F123I and L225LI induce CARD11 hyperactivity by disrupting autoinhibition by the CARD11 ID. These mutations disrupt ID-mediated intramolecular interactions and ID-dependent inhibition and bypass the requirement for ID phosphorylation during T cell receptor signaling. Intriguingly, these mutations selectively enhance the apparent affinity of CARD11 for Bcl10, but not for other signaling proteins that are recruited to CARD11 in an ID-dependent manner during normal antigen receptor signaling. Our results establish a mechanism that explains how DLBCL-associated mutations in CARD11 can initiate spontaneous, receptor-independent activation of NF-κB.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/genética , Carcinógenos , Guanilato Ciclase/genética , Mutação/genética , Proteína Quinase C/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Proteína 10 de Linfoma CCL de Células B , Humanos , Ativação Linfocitária/genética , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/metabolismo , Linfoma Difuso de Grandes Células B/patologia , NF-kappa B/genética , NF-kappa B/metabolismo , Fosforilação , Proteína Quinase C/genética , Estrutura Terciária de Proteína , Receptores de Antígenos/genética , Receptores de Antígenos/metabolismo , Transdução de Sinais/genética
17.
Immunol Res ; 47(1-3): 172-8, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20069388

RESUMO

Immunologic research has benefited tremendously from expression-cloning strategies designed to isolate genes responsible for a wide variety of immunomodulatory activities, including cytokines, receptors, signaling proteins, and transcription factors. Here, we discuss the use of expression-cloning strategies that have been modified to detect cDNAs that influence gene expression as assayed by a transcriptional reporter. We summarize our experience with these screens, review important parameters, and discuss potential modifications.


Assuntos
Clonagem Molecular/métodos , Citocinas/genética , Regulação da Expressão Gênica , Biblioteca Gênica , Fatores de Transcrição/genética , Transcrição Gênica , Linhagem Celular , Citocinas/metabolismo , Genes Reporter , HIV-1/fisiologia , Humanos , Fatores de Transcrição/metabolismo
18.
Science ; 310(5755): 1782-6, 2005 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-16357253

RESUMO

Lighter variations of pigmentation in humans are associated with diminished number, size, and density of melanosomes, the pigmented organelles of melanocytes. Here we show that zebrafish golden mutants share these melanosomal changes and that golden encodes a putative cation exchanger slc24a5 (nckx5) that localizes to an intracellular membrane, likely the melanosome or its precursor. The human ortholog is highly similar in sequence and functional in zebrafish. The evolutionarily conserved ancestral allele of a human coding polymorphism predominates in African and East Asian populations. In contrast, the variant allele is nearly fixed in European populations, is associated with a substantial reduction in regional heterozygosity, and correlates with lighter skin pigmentation in admixed populations, suggesting a key role for the SLC24A5 gene in human pigmentation.


Assuntos
Antiporters/genética , Pigmentação da Pele/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Negro ou Afro-Americano/genética , Alanina/genética , Alelos , Sequência de Aminoácidos , Animais , Antiporters/química , Antiporters/fisiologia , Povo Asiático/genética , Evolução Biológica , População Negra/genética , Cálcio/metabolismo , Frequência do Gene , Genes , Variação Genética , Haplótipos , Heterozigoto , Humanos , Transporte de Íons , Melaninas/análise , Melanossomas/química , Melanossomas/ultraestrutura , Camundongos , Dados de Sequência Molecular , Herança Multifatorial , Mutação , Epitélio Pigmentado Ocular/química , Epitélio Pigmentado Ocular/ultraestrutura , Polimorfismo de Nucleotídeo Único , Seleção Genética , Treonina/genética , População Branca/genética , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA