Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Appl Environ Microbiol ; 90(4): e0150023, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38456674

RESUMO

Plasmalogen is a specific glycerophospholipid present in both animal and bacterial organisms. It plays a crucial function in eukaryotic cellular processes and is closely related to several human diseases, including neurological disorders and cancers. Nonetheless, the precise biological role of plasmalogen in bacteria is not well understood. In this study, we identified SMU_438c as the enzyme responsible for plasmalogen production in Streptococcus mutans under anaerobic conditions. The heterologous expression of SMU_438c in a plasmalogen-negative strain, Streptococcus sanguinis, resulted in the production of plasmalogen, indicating that this enzyme is sufficient for plasmalogen production. Additionally, the plasmalogen-deficient S. mutans exhibited significantly lower acid tolerance and diminished its colonization in Drosophila flies compared to the wild-type strain and complemented strain. In summary, our data suggest that plasmalogen plays a vital role in bacterial stress tolerance and in vivo colonization. IMPORTANCE: This study sheds light on the biological role of plasmalogen, a specific glycerophospholipid, in bacteria, particularly in Streptococcus mutans. Plasmalogens are known for their significant roles in eukaryotic cells and have been linked to human diseases like neurological disorders and cancers. The enzyme SMU_438c, identified as essential for plasmalogen production under anaerobic conditions, was crucial for acid tolerance and in vivo colonization in Drosophila by S. mutans, underscoring its importance in bacterial stress response and colonization. These findings bridge the knowledge gap in bacterial physiology, highlighting plasmalogen's role in microbial survival and offering potential insights into microbial pathogenesis and host-microbe interactions.


Assuntos
Neoplasias , Doenças do Sistema Nervoso , Humanos , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Plasmalogênios/metabolismo , Streptococcus mutans/metabolismo , Ácidos/metabolismo , Drosophila , Biofilmes
2.
bioRxiv ; 2023 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-38168339

RESUMO

Early childhood dental caries (ECC) is the most common chronic disease among children with a heavy disease burden among low socioeconomic populations. Streptococcus mutans is most frequently associated with initiation of ECC. Many studies report children with multiple S. mutans strains (i.e., genotypes) having greater odds of developing ECC, studies investigating intraspecies interactions in dental caries are lacking. In this study, the impact of intraspecies interactions on cariogenic and fitness traits of clinical S. mutans isolates are investigated using in-vitro and in-vivo approaches. Initially clinical S. mutans isolates of 10 children from a longitudinal epidemiological study were evaluated. S. mutans strains (G09 and G18, most prevalent) isolated from one child were used for subsequent analysis. Association analysis was used to determine if presence of multiple S. mutans genotypes within the first-year of colonization was associated with caries. Biofilm analysis was performed for single and mixed cultures to assess cariogenic traits, including biofilm biomass, intra-polysaccharide, pH, and glucan. Confocal Laser Scanning Microscopy (CLSM) and time-lapse imaging were used to evaluate spatial and temporal biofilm dynamics, respectively. A Drosophila model was used to assess colonization in-vivo. Mean biofilm pH was significantly lower in co-cultured biofilms as compared with monoculture biofilms. Doubling of S. mutans in-vitro biofilms was observed by CLSM and in-vivo colonization in Drosophila for co-cultured S. mutans. Individual strains occupied specific domains in co-culture and G09 contributed most to increased co-culture biofilm thickness and colonization in Drosophila. Biofilm formation and acid production displayed distinct signatures in time-lapsed experiments.

3.
Front Cell Infect Microbiol ; 13: 1279380, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38192401

RESUMO

The fruit fly Drosophila melanogaster has emerged as a valuable model for investigating human biology, including the role of the microbiome in health and disease. Historically, studies involving the infection of D. melanogaster with single microbial species have yielded critical insights into bacterial colonization and host innate immunity. However, recent evidence has underscored that multiple microbial species can interact in complex ways through physical connections, metabolic cross-feeding, or signaling exchanges, with significant implications for healthy homeostasis and the initiation, progression, and outcomes of disease. As a result, researchers have shifted their focus toward developing more robust and representative in vivo models of co-infection to probe the intricacies of polymicrobial synergy and dysbiosis. This review provides a comprehensive overview of the pioneering work and recent advances in the field, highlighting the utility of Drosophila as an alternative model for studying the multifaceted microbial interactions that occur within the oral cavity and other body sites. We will discuss the factors and mechanisms that drive microbial community dynamics, as well as their impacts on host physiology and immune responses. Furthermore, this review will delve into the emerging evidence that connects oral microbes to systemic conditions in both health and disease. As our understanding of the microbiome continues to evolve, Drosophila offers a powerful and tractable model for unraveling the complex interplay between host and microbes including oral microbes, which has far-reaching implications for human health and the development of targeted therapeutic interventions.


Assuntos
Coinfecção , Drosophila melanogaster , Humanos , Animais , Disbiose , Drosophila , Cognição
4.
Am J Transl Res ; 14(6): 3729-3740, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35836842

RESUMO

OBJECTIVE: To investigate the mechanism of lncRNA MALAT1 (MALAT1) inhibiting the proliferation and invasiveness of laryngeal squamous cell carcinoma (LSCC) Hep-2 cells by modulating miR-362-3p. METHODS: We collected the expression profile of lncRNAs and miRNAs in LSCC downloaded from The Cancer Genome Atlas (TCGA) database as well as LSCC tissue samples and adjacent normal counterparts resected from LSCC patients in Lvliang People's Hospital and First Hospital of Shanxi Medical University between January 2018 and June 2020 for analysis. Human LSCC Hep-2 cells were selected for experiments. The expression of miR-362-3p and MALAT1 was detected by quantitative real-time polymerase chain reaction (qRT-PCR). Cells were subsequently transfected to knock out MALAT1, and the growth, metastasis and invasiveness of cells were evaluated by CCK-8 assay, plate clone formation, wound healing, and Transwell invasion assays respectively. The binding of MALAT1 to miR-362-3p was verified by RNA pull-down, RNA binding protein immunoprecipitation (RIP), and dual-luciferase reporter assays. RESULTS: MALAT1 was highly expressed while miR-362-3p was lowly expressed in both LSCC tissues and cells compared with normal counterparts. MALAT1 knockdown inhibited the viability of Hep-2 cells, reducing the number of plate clone-forming cells as well as the number of migrated and invaded cells. Transfection of miR-362-3p inhibitor into Hep-2 cells treated by si-MALAT1 reversed the inhibition of si-MALAT1 on the proliferation of Hep-2 cells, and promoted cell invasiveness and migration. MALAT1 can sponge miR-362-3p and inhibit its expression. CONCLUSIONS: Knockdown of MALAT can inhibit Hep-2 cell proliferation and reduce its invasiveness and migration by modulating miR-362-3p.

5.
PLoS Genet ; 17(11): e1009894, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34748546

RESUMO

The generation of a diversity of photoreceptor (PR) subtypes with different spectral sensitivities is essential for color vision in animals. In the Drosophila eye, the Hippo pathway has been implicated in blue- and green-sensitive PR subtype fate specification. Specifically, Hippo pathway activation promotes green-sensitive PR fate at the expense of blue-sensitive PRs. Here, using a sensitized triple heterozygote-based genetic screening approach, we report the identification of the single Drosophila zonula occludens-1 (ZO-1) protein Polychaetoid (Pyd) as a new regulator of the Hippo pathway during the blue- and green-sensitive PR subtype binary fate choice. We demonstrate that Pyd acts upstream of the core components and the upstream regulator Pez in the Hippo pathway. Furthermore, We found that Pyd represses the activity of Su(dx), a E3 ligase that negatively regulates Pez and can physically interact with Pyd, during PR subtype fate specification. Together, our results identify a new mechanism underlying the Hippo signaling pathway in post-mitotic neuronal fate specification.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Junções Íntimas/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo , Animais , Proteínas de Drosophila/genética , Heterozigoto , Transdução de Sinais , Proteínas de Junções Íntimas/genética , Proteína da Zônula de Oclusão-1/genética
6.
Dev Biol ; 455(1): 51-59, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31265830

RESUMO

The Hippo tumor suppressor pathway plays many fundamental cell biological roles during animal development. Two central players in controlling Hippo-dependent gene expression are the TEAD transcription factor Scalloped (Sd) and its transcriptional co-activator Yorkie (Yki). Hippo signaling phosphorylates Yki, thereby blocking Yki-dependent transcriptional control. In post-mitotic Drosophila photoreceptors, a bistable negative feedback loop forms between the Hippo-dependent kinase Warts/Lats and Yki to lock in green vs blue-sensitive neuronal subtype choices, respectively. Previous experiments indicate that sd and yki mutants phenocopy each other's functions, both being required for promoting the expression of the blue photoreceptor fate determinant melted (melt) and the blue-sensitive opsin Rh5. Here, we demonstrate that Sd ensures the robustness of this neuronal fate decision via multiple antagonistic gene regulatory roles. In Hippo-positive (green) photoreceptors, Sd directly represses both melt and Rh5 gene expression through defined TEAD binding sites, a mechanism that is antagonized by Yki in Hippo-negative (blue) cells. Additionally, in blue photoreceptors, Sd is required to promote the translation of the Rh5 protein through a 3'UTR-dependent and microRNA-mediated process. Together, these studies reveal that Sd can drive context-dependent cell fate decisions through opposing transcriptional and post-transcriptional mechanisms.


Assuntos
Proteínas de Drosophila/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neurônios/metabolismo , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinases/genética , Processamento Pós-Transcricional do RNA , Transativadores/genética , Fatores de Transcrição/genética , Transcrição Gênica , Animais , Diferenciação Celular/genética , Linhagem Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurônios/citologia , Proteínas Nucleares/metabolismo , Células Fotorreceptoras de Invertebrados/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Rodopsina/genética , Rodopsina/metabolismo , Transdução de Sinais/genética , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Sinalização YAP
7.
Science ; 342(6155): 1238016, 2013 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-23989952

RESUMO

Signaling pathways are reused for multiple purposes in plant and animal development. The Hippo pathway in mammals and Drosophila coordinates proliferation and apoptosis via the coactivator and oncoprotein YAP/Yorkie (Yki), which is homeostatically regulated through negative feedback. In the Drosophila eye, cross-repression between the Hippo pathway kinase LATS/Warts (Wts) and growth regulator Melted generates mutually exclusive photoreceptor subtypes. Here, we show that this all-or-nothing neuronal differentiation results from Hippo pathway positive feedback: Yki both represses its negative regulator, warts, and promotes its positive regulator, melted. This postmitotic Hippo network behavior relies on a tissue-restricted transcription factor network-including a conserved Otx/Orthodenticle-Nrl/Traffic Jam feedforward module-that allows Warts-Yki-Melted to operate as a bistable switch. Altering feedback architecture provides an efficient mechanism to co-opt conserved signaling networks for diverse purposes in development and evolution.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurogênese/genética , Proteínas Nucleares/metabolismo , Células Fotorreceptoras de Invertebrados/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Retroalimentação Fisiológica , Redes Reguladoras de Genes , Proteínas de Homeodomínio/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mitose , Células Fotorreceptoras de Invertebrados/citologia , Células Fotorreceptoras de Invertebrados/metabolismo , Proteínas Quinases/genética , Proteínas de Sinalização YAP
8.
Dev Dyn ; 241(1): 215-28, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22113834

RESUMO

BACKGROUND: Otd-related transcription factors are evolutionarily conserved to control anterior patterning and neurogenesis. In humans, two such factors, OTX2 and CRX, are expressed in all photoreceptors from early specification through adulthood and associate with several photoreceptor-specific retinopathies. It is not well understood how these factors function independently vs. redundantly, or how specific mutations lead to different disease outcomes. It is also unclear how OTX1 and OTX2 functionally overlap during other aspects of neurogenesis and ocular development. Drosophila encodes a single Otd factor that has multiple functions during eye development. Using the Drosophila eye as a model, we tested the ability of the human OTX1, OTX2, and CRX genes, as well as several disease-associated CRX alleles, to rescue the different functions of Otd. RESULTS: Our results indicate the following: OTX2 and CRX display overlapping, yet distinct subfunctions of Otd during photoreceptor differentiation; CRX disease alleles can be functionally distinguished based on their rescue properties; and all three factors are able to rescue rhabdomeric photoreceptor morphogenesis. CONCLUSIONS: Our findings have important implications for understanding how Otx proteins have subfunctionalized during evolution, and cement Drosophila as an effective tool to unravel the molecular bases of photoreceptor pathogenesis.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/fisiologia , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição Otx/metabolismo , Transativadores/metabolismo , Animais , Diferenciação Celular/fisiologia , Proteínas de Drosophila/genética , Proteínas de Homeodomínio/genética , Humanos , Morfogênese/fisiologia , Neurogênese/fisiologia , Fatores de Transcrição Otx/genética , Células Fotorreceptoras de Invertebrados/citologia , Células Fotorreceptoras de Invertebrados/fisiologia , Transativadores/genética
9.
Cell ; 145(6): 956-68, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21663797

RESUMO

How complex networks of activators and repressors lead to exquisitely specific cell-type determination during development is poorly understood. In the Drosophila eye, expression patterns of Rhodopsins define at least eight functionally distinct though related subtypes of photoreceptors. Here, we describe a role for the transcription factor gene defective proventriculus (dve) as a critical node in the network regulating Rhodopsin expression. dve is a shared component of two opposing, interlocked feedforward loops (FFLs). Orthodenticle and Dve interact in an incoherent FFL to repress Rhodopsin expression throughout the eye. In R7 and R8 photoreceptors, a coherent FFL relieves repression by Dve while activating Rhodopsin expression. Therefore, this network uses repression to restrict and combinatorial activation to induce cell-type-specific expression. Furthermore, Dve levels are finely tuned to yield cell-type- and region-specific repression or activation outcomes. This interlocked FFL motif may be a general mechanism to control terminal cell-fate specification.


Assuntos
Proteínas de Drosophila/genética , Drosophila/embriologia , Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Células Fotorreceptoras de Invertebrados/metabolismo , Rodopsina/genética , Animais , Drosophila/citologia , Proteínas de Drosophila/metabolismo , Olho/embriologia , Retroalimentação Fisiológica , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/metabolismo
10.
Neural Dev ; 6: 20, 2011 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-21539742

RESUMO

BACKGROUND: The concept of an equivalence group, a cluster of cells with equal potential to adopt the same specific fate, has served as a useful paradigm to understand neural cell type specification. In the Drosophila eye, a set of five cells, called the 'R7 equivalence group', generates a single photoreceptor neuron and four lens-secreting epithelial cells. This choice between neuronal versus non-neuronal cell fates rests on differential requirements for, and cross-talk between, Notch/Delta- and Ras/mitogen-activated protein kinase (MAPK)-dependent signaling pathways. However, many questions remain unanswered related to how downstream events of these two signaling pathways mediate distinct cell fate decisions. RESULTS: Here, we demonstrate that two direct downstream targets of Ras and Notch signaling, the transcription factors Prospero and dPax2, are essential regulators of neuronal versus non-neuronal cell fate decisions in the R7 equivalence group. Prospero controls high activated MAPK levels required for neuronal fate, whereas dPax2 represses Delta expression to prevent neuronal fate. Importantly, activity from both factors is required for proper cell fate decisions to occur. CONCLUSIONS: These data demonstrate that Ras and Notch signaling are integrated during cell fate decisions within the R7 equivalence group through the combinatorial and opposing activities of Pros and dPax2. Our study provides one of the first examples of how the differential expression and synergistic roles of two independent transcription factors determine cell fate within an equivalence group. Since the integration of Ras and Notch signaling is associated with many developmental and cancer models, these findings should provide new insights into how cell specificity is achieved by ubiquitously used signaling pathways in diverse biological contexts.


Assuntos
Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/fisiologia , Proteínas Nucleares/fisiologia , Fator de Transcrição PAX2/fisiologia , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/fisiologia , Proteínas ras/metabolismo , Animais , Animais Geneticamente Modificados , Diferenciação Celular/genética , Drosophila , Proteínas de Drosophila/genética , Olho/citologia , Olho/embriologia , Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas do Tecido Nervoso/genética , Neurônios/classificação , Proteínas Nucleares/genética , Fator de Transcrição PAX2/genética , Células Fotorreceptoras , Pupa , Receptores Notch/genética , Retina/citologia , Células Fotorreceptoras Retinianas Cones/citologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Fatores de Transcrição/genética , Proteínas ras/genética
11.
Dev Biol ; 347(1): 122-32, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20732315

RESUMO

Orthodenticle (Otd)-related transcription factors are essential for anterior patterning and brain morphogenesis from Cnidaria to Mammals, and genetically underlie several human retinal pathologies. Despite their key developmental functions, relatively little is known regarding the molecular basis of how these factors regulate downstream effectors in a cell- or tissue-specific manner. Many invertebrate and vertebrate species encode two to three Otd proteins, whereas Drosophila encodes a single Otd protein. In the fly retina, Otd controls rhabdomere morphogenesis of all photoreceptors and regulates distinct Rhodopsin-encoding genes in a photoreceptor subtype-specific manner. Here, we performed a structure-function analysis of Otd during Drosophila eye development using in vivo rescue experiments and in vitro transcriptional regulatory assays. Our findings indicate that Otd requires at least three distinct transcriptional regulatory domains to control photoreceptor-specific rhodopsin gene expression and photoreceptor morphogenesis. Our results also uncover a previously unknown role for Otd in preventing co-expression of sensory receptors in blue vs. green-sensitive R8 photoreceptors. Sequence analysis indicates that many of the transcriptional regulatory domains identified here are conserved in multiple Diptera Otd-related proteins. Thus, these studies provide a basis for identifying shared molecular pathways involved in a wide range of developmental processes.


Assuntos
Diferenciação Celular/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Proteínas de Homeodomínio/genética , Células Fotorreceptoras de Invertebrados/citologia , Sequências Reguladoras de Ácido Nucleico/genética , Transcrição Gênica , Sequência de Aminoácidos , Animais , Sequência Conservada/genética , Proteínas de Drosophila/química , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/química , Dados de Sequência Molecular , Morfogênese/genética , Células Fotorreceptoras de Invertebrados/metabolismo , Regiões Promotoras Genéticas/genética , Rodopsina/genética , Rodopsina/metabolismo , Alinhamento de Sequência , Deleção de Sequência/genética , Homologia de Sequência de Aminoácidos
12.
Development ; 134(23): 4243-53, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17978002

RESUMO

A major question in development is how different specialized cell types arise from a common progenitor. In the adult Drosophila compound eye, color discrimination is achieved by UV-, blue- and green-sensitive photoreceptors (PRs). These different PR subsets arise from neuronal precursors called R7 and R8 cells. Recent studies have demonstrated that R7-based UV-sensitive PRs require the repression of R8-based blue/green-sensitive PR characteristics to properly develop. This repression is mediated by the transcription factor Prospero (Pros). Here, we report that Senseless (Sens), a Drosophila ortholog of the vertebrate Gfi1 transcription factor, plays an opposing role to Pros by both negatively regulating R7-based features and positively enforcing R8-based features during terminal differentiation. In addition, we demonstrate that Pros and Sens function together with the transcription factor Orthodenticle (Otd) to oppositely regulate R7 and R8 PR Rhodopsin gene expression in vitro. These data show that sens, previously shown to be essential for neuronal specification, also controls differentiation of specific neuronal subtypes in the retina. Interestingly, Pros has recently been shown to function as a tumor suppressor, whereas Gfi1 is a well-characterized oncogene. Thus, we propose that sens/pros antagonism is important for regulating many biological processes.


Assuntos
Percepção de Cores/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila/fisiologia , Proteínas Nucleares/fisiologia , Células Fotorreceptoras de Invertebrados/fisiologia , Fatores de Transcrição/fisiologia , Animais , Cromossomos/genética , Drosophila/genética , Proteínas de Drosophila/genética , Imunofluorescência , Proteínas Nucleares/genética , Células Fotorreceptoras de Invertebrados/citologia , Regiões Promotoras Genéticas , Opsinas de Bastonetes/antagonistas & inibidores , Opsinas de Bastonetes/genética , Fatores de Transcrição/genética
13.
Acta Biochim Biophys Sin (Shanghai) ; 38(9): 625-32, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16953301

RESUMO

Using purified recombinant human ventricular myosin light chain 1 (HVMLC1) as the antigen, three monoclonal antibodies, designated C8, C9 and B12, were prepared. Immunoblot experiments demonstrated that all monoclonal antibodies could react with the ventricular myosin light chain 1 isolated from different sources, such as human, rat or pig. It was also demonstrated that C8 was directed against the NN part of the N-fragment (amino acid 1-40) of HVMLC1, and both C9 and B12 against the C-fragment (amino acid 99-195). The affinity constants of C8, C9 and B12 were 3.20 x 10(8), 8.600 x 10(7) and 1.770 x 10(8) M(-1), respectively, determined by non-competitive ELISA. The isotype of B12 was determined as IgG2a, whereas the isotype of both C8 and C9 were IgG1. In the presence of C9 or B12, the actin-activated Mg(2+)ATPase activity of myosin was greatly inhibited, but there was almost no effect on the Mg(2+) ATPase activity for C8. B12 and C9 also inhibited the superprecipitation of porcine cardiac native actomyosin (myosin B) and reconstituted actomyosin, but C8 did not. The results indicate that all three monoclonal antibodies could bind the intact myosin molecule, but B12 and C9 might more easily react with epitopes located in the C-fragment of HVMLC1. The inhibitory effects of B12 and C9 on ATPase activity and superprecipitation assays show that light chain 1, particularly the C-fragment domain, is involved in the modulation of the actin-activated Mg(2+) ATPase activity of myosin and, as a consequence, plays an essential role in the interaction of actin and myosin.


Assuntos
Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , ATPase de Ca(2+) e Mg(2+)/imunologia , Ventrículos do Coração/imunologia , Cadeias Leves de Miosina/imunologia , Engenharia de Proteínas/métodos , Animais , Células Cultivadas , Humanos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/imunologia , Suínos
14.
Biophys Chem ; 106(1): 57-66, 2003 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-14516912

RESUMO

The biological functions of the myosin light chain 1 (LC1) have not been clearly elucidated yet. In this work we cloned and expressed N- and C- terminal fragments of human ventricular LC1 (HVLC1) containing amino acid residues 1-98 and 99-195 and two parts, NN and NC of N fragment in GST-fusion forms, respectively. Using GST pull-down assay, the direct binding experiments of LC1 with rat cardiac G-actin, F-actin and thin filaments, as well as rat cardiac myosin heavy chain (RCMHC) have been performed. Furthermore, the recombinant complexes of rat myosin S1 with N- and C-fragments, as well as the whole molecular of HVLC1 were generated. The results suggested that both binding sites of HVLC1 for actin and myosin heavy chain are positioned in its N-terminal fragment, which may contain several actin-binding sites in tandem. The polymerization of G-actin, the tropomyosin and troponin molecules located in the thin filaments do not hinder the binding of N-terminal fragment of HVLC1 with actin and thin filaments in vitro. The recombinant complex of rat cardiac myosin S1 (RCMS1) with N fragment of HVLC1 greatly decreased actin-activated Mg(2+)-ATPase activity for lack of C fragment. We conclude that the N-fragment is the binding domain of human ventricular LC1, whereas the C-fragment serves as a functional domain, which may be more involved in the modulation of the actin-activated ATPase activity of myosin.


Assuntos
Ventrículos do Coração/química , Cadeias Leves de Miosina/química , Cadeias Leves de Miosina/metabolismo , Actinas/metabolismo , Adenosina Trifosfatases/química , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Cálcio/metabolismo , Humanos , Magnésio/metabolismo , Miocárdio/química , Cadeias Leves de Miosina/genética , Subfragmentos de Miosina , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Potássio/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Ratos
15.
Artigo em Inglês | MEDLINE | ID: mdl-12796811

RESUMO

The genome of wheat rosette stunt virus (WRSV), a plant rhabdovirus, is a single negative strand RNA. It encodes five viral structural proteins: the glycoprotein (G), the matrix protein (M), the nucleocapsid protein (N), the large protein (L) and the non?structural protein (NS), which was later proved to be a viral structural protein too and existed in a variety of phosphorylation forms in case of vascular stomatitis virus (VSV). In this paper we demonstrated that NS protein of WRSV, either bound with the viral nucleocapsid or expressed in bacteria could be in vitro phosphorylated in presence of viral nucleocapsid. We concluded that the NS protein of WRSV was a phosphorylated protein and it might exist in both phosphorylated and dephosphorylated forms in virions. Our results excluded the possibility that the NS protein could be autophosphorylated. The L protein, the major component of viral RNA dependent RNA polymerase is associated with the protein kinase for phosphorylation of NS protein.


Assuntos
Nucleocapsídeo/metabolismo , Vírus de Plantas/metabolismo , Proteínas não Estruturais Virais/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Nucleocapsídeo/isolamento & purificação , Nucleocapsídeo/ultraestrutura , Fosforilação , Vírus de Plantas/química , Vírus de Plantas/genética , Proteínas Quinases/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/isolamento & purificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA