Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 17162, 2023 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-37821496

RESUMO

The immunoglobulin (Ig) superfamily members are involved in cell adhesion and migration, complex multistep processes that play critical roles in embryogenesis, wound healing, tissue formation, and many other processes, but their specific functions during embryonic development remain unclear. Here, we have studied the function of the immunoglobulin superfamily member 3 (IGSF3) by generating an Igsf3 knockout (KO) mouse model with CRISPR/Cas9-mediated genome engineering. By combining RNA and protein detection methodology, we show that during development, IGSF3 localizes to the neural crest and a subset of its derivatives, suggesting a role in normal embryonic and early postnatal development. Indeed, inactivation of Igsf3 impairs the ability of the vagal neural crest cells to migrate and normally innervate the intestine. The small intestine of Igsf3 KO mice shows reduced thickness of the muscularis externa and diminished number of enteric neurons. Also, misalignment of neurons and smooth muscle cells in the developing intestinal villi is detected. Taken together, our results suggest that IGSF3 functions contribute to the formation of the enteric nervous system. Given the essential role of the enteric nervous system in maintaining normal gastrointestinal function, our study adds to the pool of information required for further understanding the mechanisms of gut innervation and etiology behind bowel motility disorders.


Assuntos
Sistema Nervoso Entérico , Crista Neural , Camundongos , Animais , Neurônios/fisiologia , Trato Gastrointestinal , Sistema Nervoso Entérico/metabolismo , Intestino Delgado , Imunoglobulinas/genética , Imunoglobulinas/metabolismo , Movimento Celular/fisiologia
2.
Mol Cancer Ther ; 22(11): 1319-1331, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37486980

RESUMO

Chimeric antigen receptor (CAR) T-cell immunotherapies for solid tumors face critical challenges such as heterogeneous antigen expression. We characterized stage-specific embryonic antigen-4 (SSEA-4) cell-surface glycolipid as a target for CAR T-cell therapy. SSEA-4 is mainly expressed during embryogenesis but is also found in several cancer types making it an attractive tumor-associated antigen. Anti-SSEA-4 CAR-T cells were generated and assessed preclinically in vitro and in vivo for antitumor response and safety. SSEA-4 CAR-T cells effectively eliminated SSEA-4-positive cells in all the tested cancer cell lines, whereas SSEA-4-negative cells lines were not targeted. In vivo efficacy and safety studies using NSG mice and the high-grade serous ovarian cancer cell line OVCAR4 demonstrated a remarkable and specific antitumor response at all the CAR T-cell doses used. At high T-cell doses, CAR T cell-treated mice showed signs of health deterioration after a follow-up period. However, the severity of toxicity was reduced with a delayed onset when lower CAR T-cell doses were used. Our data demonstrate the efficacy of anti-SSEA-4 CAR T-cell therapy; however, safety strategies, such as dose-limiting and/or equipping CAR-T cells with combinatorial antigen recognition should be implemented for its potential clinical translation.


Assuntos
Carcinoma , Neoplasias Ovarianas , Receptores de Antígenos Quiméricos , Humanos , Feminino , Animais , Camundongos , Glicoesfingolipídeos/metabolismo , Linhagem Celular Tumoral , Neoplasias Ovarianas/metabolismo , Imunoterapia Adotiva , Linfócitos T , Carcinoma Epitelial do Ovário/metabolismo , Carcinoma/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Int J Mol Sci ; 24(7)2023 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-37047272

RESUMO

After seeing a dramatic increase in the development and use of immunotherapy and precision medicine over the past few decades, oncological care now embraces the start of the adoptive cell therapy (ACT) era. This impulse towards a new treatment paradigm has been led by chimeric antigen receptor (CAR) T cells, the only type of ACT medicinal product to be commercialized so far. Brought about by an ever-growing understanding of cellular engineering, CAR T cells are T lymphocytes genetically modified with an appropriate DNA construct, which endows them with expression of a CAR, a fusion protein between a ligand-specific recognition domain, often an antibody-like structure, and the activating signaling domain of the T cell receptor. Through this genetic enhancement, CAR T cells are engineered from a cancer patient's own lymphocytes to better target and kill their cancer cells, and the current amassed data on clinical outcomes point to a stream of bright developments in the near future. Herein, from concept design and present-day manufacturing techniques to pressing hurdles and bright discoveries around the corner, we review and thoroughly describe the state of the art in CAR T cell therapy.


Assuntos
Imunoterapia Adotiva , Neoplasias , Humanos , Imunoterapia Adotiva/métodos , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T , Neoplasias/terapia
4.
mBio ; 14(1): e0322122, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36656016

RESUMO

Rapid phagosomal escape mediated by listeriolysin O (LLO) is a prerequisite for Listeria monocytogenes intracellular replication and pathogenesis. Escape takes place within minutes after internalization from vacuoles that are negative to the early endosomal Rab5 GTPase and positive to the late endosomal Rab7. Using mutant analysis, we found that the listerial invasin InlB was required for optimal intracellular proliferation of L. monocytogenes. Starting from this observation, we determined in HeLa cells that InlB promotes early phagosomal escape and efficient Rab7 acquisition by the Listeria-containing vacuole (LCV). Recruitment of the class III phosphoinositide 3-kinase (PI3K) Vps34 to the LCV and accumulation of its lipid product, phosphatidylinositol 3-phosphate (PI3P), two key endosomal maturation mediators, were also dependent on InlB. Small interfering RNA (siRNA) knockdown experiments showed that Vps34 was required for Rab7 recruitment and early (LLO-mediated) escape and supported InlB-dependent intracellular proliferation. Together, our data indicate that InlB accelerates LCV conversion into an escape-favorable Rab7 late phagosome via subversion of class III PI3K/Vps34 signaling. Our findings uncover a new function for the InlB invasin in Listeria pathogenesis as an intracellular proliferation-promoting virulence factor. IMPORTANCE Avoidance of lysosomal killing by manipulation of the endosomal compartment is a virulence mechanism assumed to be largely restricted to intravacuolar intracellular pathogens. Our findings are important because they show that cytosolic pathogens like L. monocytogenes, which rapidly escape the phagosome after internalization, can also extensively subvert endocytic trafficking as part of their survival strategy. They also clarify that, instead of delaying phagosome maturation (to allow time for LLO-dependent disruption, as currently thought), via InlB L. monocytogenes appears to facilitate the rapid conversion of the phagocytic vacuole into an escape-conducive late phagosome. Our data highlight the multifunctionality of bacterial virulence factors. At the cell surface, the InlB invasin induces receptor-mediated phagocytosis via class I PI3K activation, whereas after internalization it exploits class III PI3K (Vsp34) to promote intracellular survival. Systematically elucidating the mechanisms by which Listeria interferes with PI3K signaling all along the endocytic pathway may lead to novel anti-infective therapies.


Assuntos
Listeria monocytogenes , Listeria , Humanos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proliferação de Células , Células HeLa , Proteínas Hemolisinas/genética , Fosfatidilinositol 3-Quinases/metabolismo , Vacúolos/metabolismo , Classe III de Fosfatidilinositol 3-Quinases
5.
Exp Cell Res ; 355(1): 26-39, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28341445

RESUMO

Cellular interactions with the extracellular environment are modulated by cell surface polysialic acid (PSA) carried by the neural cell adhesion molecule (NCAM). PSA-NCAM is involved in cellular processes such as differentiation, plasticity, and migration, and is elevated in Alzheimer's disease as well as in metastatic tumour cells. Our previous work demonstrated that insulin enhances the abundance of cell surface PSA by inhibiting PSA-NCAM endocytosis. In the present study we have identified a mechanism for insulin-dependent inhibition of PSA-NCAM turnover affecting cell migration. Insulin enhanced the phosphorylation of the focal adhesion kinase leading to dissociation of αv-integrin/PSA-NCAM clusters, and promoted cell migration. Our results show that αv-integrin plays a key role in the PSA-NCAM turnover process. αv-integrin knockdown stopped PSA-NCAM from being endocytosed, and αv-integrin/PSA-NCAM clusters co-labelled intracellularly with Rab5, altogether indicating a role for αv-integrin as a carrier for PSA-NCAM during internalisation. Furthermore, inhibition of p-FAK caused dissociation of αv-integrin/PSA-NCAM clusters and counteracted the insulin-induced accumulation of PSA at the cell surface and cell migration was impaired. Our data reveal a functional association between the insulin/p-FAK-dependent regulation of PSA-NCAM turnover and cell migration through the extracellular matrix. Most importantly, they identify a novel mechanism for insulin-stimulated cell migration.


Assuntos
Movimento Celular/efeitos dos fármacos , Insulina/farmacologia , Moléculas de Adesão de Célula Nervosa/metabolismo , Ácidos Siálicos/antagonistas & inibidores , Animais , Bovinos , Relação Dose-Resposta a Droga , Humanos , Insulina/química , Pâncreas/química , Ácidos Siálicos/metabolismo , Relação Estrutura-Atividade , Células Tumorais Cultivadas
6.
Sci Rep ; 6: 26587, 2016 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-27215737

RESUMO

The human brain is a highly vascular organ in which the blood-brain barrier (BBB) tightly regulates molecules entering the brain. Pericytes are an integral cell type of the BBB, regulating vascular integrity, neuroinflammation, angiogenesis and wound repair. Despite their importance, identifying pericytes amongst other perivascular cell types and deciphering their specific role in the neurovasculature remains a challenge. Using primary adult human brain cultures and fluorescent-activated cell sorting, we identified two CD73(+)CD45(-) mesenchymal populations that showed either high or low CD90 expression. CD90 is known to be present on neurons in the brain and peripheral blood vessels. We found in the human brain, that CD90 immunostaining localised to the neurovasculature and often associated with pericytes. In vitro, CD90(+) cells exhibited higher basal proliferation, lower expression of markers αSMA and CD140b, produced less extracellular matrix (ECM) proteins, and exhibited lesser pro-inflammatory responses when compared to the CD90(-) population. Thus, CD90 distinguishes two interrelated, yet functionally distinct pericyte populations in the adult human brain that may have discrete roles in neurovascular function, immune response and scar formation.


Assuntos
Encéfalo/citologia , Pericitos/citologia , Antígenos Thy-1/metabolismo , Adulto , Biomarcadores/metabolismo , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Proliferação de Células , Células Cultivadas , Feminino , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Masculino , Pericitos/metabolismo , Fenótipo , Adulto Jovem
7.
J Neurochem ; 126(6): 758-70, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23844825

RESUMO

Cellular interactions mediated by the neural cell adhesion molecule (NCAM) are critical in cell migration, differentiation and plasticity. Switching of the NCAM-interaction mode, from adhesion to signalling, is determined by NCAM carrying a particular post-translational modification, polysialic acid (PSA). Regulation of cell-surface PSA-NCAM is traditionally viewed as a direct consequence of polysialyltransferase activity. Taking advantage of the polysialyltransferase Ca²âº-dependent activity, we demonstrate in TE671 cells that downregulation of PSA-NCAM synthesis constitutes a necessary but not sufficient condition to reduce cell-surface PSA-NCAM; instead, PSA-NCAM turnover required internalization of the molecule into the cytosol. PSA-NCAM internalization was specifically triggered by collagen in the extracellular matrix (ECM) and prevented by insulin-like growth factor (IGF1) and insulin. Our results pose a novel role for IGF1 and insulin in controlling cell migration through modulation of PSA-NCAM turnover at the cell surface. Neural cell adhesion molecules (NCAMs) are critically involved in cell differentiation and migration. Polysialylation (PSA)/desialylation of NCAMs switches their functional interaction mode and, in turn, migration and differentiation. We have found that the desialylation process of PSA-NCAM occurs via endocytosis, induced by collagen-IV and blocked by insulin-like growth factor (IGF1) and insulin, suggesting a novel association between PSA-NCAM, IGF1/insulin and brain/tumour plasticity.


Assuntos
Matriz Extracelular/metabolismo , Hipoglicemiantes/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Insulina/farmacologia , Moléculas de Adesão de Célula Nervosa/metabolismo , Ácidos Siálicos/metabolismo , Western Blotting , Cálcio/metabolismo , Linhagem Celular Tumoral , Colágeno Tipo IV/metabolismo , Endocitose/efeitos dos fármacos , Humanos , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Processamento de Proteína Pós-Traducional , Reação em Cadeia da Polimerase em Tempo Real
8.
J Chem Neuroanat ; 52: 58-68, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23770130

RESUMO

It is now well established that the human brain continuously produces new stem cells until well into old age. One of these stem-cell rich areas in the human brain is the sub-ventricular zone (SVZ). The human SVZ is organized in four distinctive layers containing type A, B and C cells. To date, no studies have investigated the distribution of inhibitory neurotransmitters such as γ-aminobutyric acid (GABA) and their respective receptors on the different cell types in the human SVZ. GABA(A) receptors (GABA(A)R) are ubiquitously expressed, inhibitory heteropentameric chloride ion channels comprised of a variety of subunits that are targeted by many prescribed drugs. In this study we present detailed immunohistochemical data on the regional and cellular localization of α1, α2, α3, ß2,3 and γ2 subunits of GABA(A)R in the human SVZ. The results from our double and triple labeling studies demonstrate that the cell types and subunit composition throughout the SVZ is heterogeneous; the thickness of the SVZ and GABA(A)R α2 and γ2 expression is increased especially in the vicinity of large SVZ blood vessels. GABA(A)R γ2 is the most specific to the SVZ and present on various cells that express, either glial fibrillary acidic protein (GFAPδ) or polysialic acid-neural cell adhesion molecule (PSA-NCAM) separately, or together in a respective ratio of 7:6:2. Proliferating (type C) cells in the SVZ express GAD65/67, GFAPδ and GABA(A)R ß2,3 receptor subunits. Within the SVZ the majority of cells have an unexpected nuclear GABA(A)R ß2,3 expression that is inversely proportional to that of PCNA (proliferating cell nuclear antigen marker), which is a very different pattern of expression compared with underlying caudate nucleus cells. Taken together our results provide a detailed description of the chemo-architecture of the adult human SVZ demonstrating the importance of GABA and GABA(A) receptors on the various cell types in the SVZ.


Assuntos
Ventrículos Cerebrais/química , Células-Tronco Neurais/química , Subunidades Proteicas/metabolismo , Receptores de GABA-A/metabolismo , Adulto , Idoso , Proliferação de Células , Ventrículos Cerebrais/citologia , Ventrículos Cerebrais/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Células-Tronco Neurais/metabolismo , Subunidades Proteicas/biossíntese , Receptores de GABA-A/biossíntese
9.
J Neurosci Methods ; 204(1): 87-103, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22101194

RESUMO

P19 embryonal carcinoma (EC) cells are an invaluable tool for approximating the mechanisms that govern neuronal differentiation but with an enormous degree of simplification and have primarily been used to model the early stages of neurogenesis. However, they are often cultured under conditions that promote unrestricted non-neuronal growth that compromises neuronal viability. In this study we report an improved method to differentiate P19 EC cells that gives rise to high yields of functionally and morphologically mature neurons while significantly reducing the over-growth of non-neuronal cells in the cultures. In this protocol, P19 EC cells are induced in Minimum Essential Medium alpha supplemented with all-trans retinoic acid (RA) and 2.5% serum, and cultured as a monolayer. After RA-induction, cells are cultured on Matrigel coated-plates using defined media comprised of Neurobasal-A medium temporally supplemented with N2 and then B-27 for the remaining culture period. By treating the culture with Cytosine ß-d-arabinofuranoside and 2'-Deoxycytidine for five days, the cultures are reliably promoted toward the neuronal differentiation vs non-neuronal differentiation, this accounting for a progressive neuronal enrichment of the cultures reaching 56% after 20 days of culture. P19-derived neural progenitor cells progressively expressed neuronal markers such as NeuN, Calretinin, Calbindin and Synapsin I in close resemblance to that occurring in vivo in the central nervous system (CNS). Furthermore, RA-induced P19 EC cells progressively acquired functional neuronal traits and after approximately 3 weeks in culture revealed mature neurophysiological properties, characteristics of CNS neurons. This protocol allows for a more specific assessment of the neuronal differentiation processes in vitro.


Assuntos
Carcinoma Embrionário/patologia , Carcinoma Embrionário/fisiopatologia , Técnicas de Cultura de Células/métodos , Neurogênese/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Tretinoína/farmacologia , Animais , Diferenciação Celular , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Camundongos
10.
Prog Brain Res ; 175: 33-42, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19660647

RESUMO

In the mammalian brain, olfaction is an important sense that is used to detect odors of different kinds that can warn of off food, to produce a mothering instinct in a flock or group of animals, and to warn of danger such as fire or poison. The olfactory system is made up of a long-distance rostral migratory stream that arises from the subventricular zone in the wall of the lateral ventricle, mainly comprises neuroblasts, and stretches all the way through the basal forebrain to terminate in the olfactory bulb. The olfactory bulb receives a constant supply of new neurons that allow ongoing integration of new and different smells, and these are integrated into either the granule cell layer or the periglomerular layer. The continuous turnover of neurons in the olfactory bulb allows us to study the proliferation, migration, and differentiation of neurons and their application in therapies for neurodegenerative diseases. In this chapter, we will examine the notion that the olfactory system might be the route of entry for factors that cause or contribute to neurodegeneration in the central nervous system. We will also discuss the enzymes that may be involved in the addition of polysialic acid to neural cell adhesion molecule, which is vital for allowing the neuroblasts to move through the rostral migratory stream. Finally, we will discuss a possible role of endosialidases for removing polysialic acid from neural cell adhesion molecules, which causes neuroblasts to stop migrating and terminally differentiate into olfactory bulb interneurons.


Assuntos
Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Neurônios/citologia , Bulbo Olfatório/citologia , Condutos Olfatórios/citologia , Células-Tronco/citologia , Animais , Encefalopatias/fisiopatologia , Humanos , Neurônios/fisiologia , Bulbo Olfatório/fisiologia , Condutos Olfatórios/fisiologia , Olfato/fisiologia , Células-Tronco/fisiologia
11.
Microbes Infect ; 9(10): 1196-207, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17764998

RESUMO

The PrfA protein, a member of the Crp/Cap-Fnr family of bacterial transcription factors, controls the expression of key virulence determinants of the facultative intracellular pathogen Listeria monocytogenes. Each of the steps of the listerial intracellular infection cycle-host cell invasion, phagosomal escape, cytosolic replication, and direct cell-to-cell spread-is mediated by products of the PrfA regulon. Only 10 of the 2853 genes of the L. monocytogenes EGDe genome have been confirmed as bona fide (directly regulated) members of this regulon, a number surprisingly small given the apparent complexity of listerial intracellular parasitism. PrfA activates transcription by binding as a dimer to a palindromic promoter element of canonical sequence tTAACanntGTtAa, with seven invariant nucleotides (in capitals) and a two-mismatch tolerance. PrfA integrates a number of environmental and bacteria-derived signals to ensure the correct spatio-temporal and niche-adapted expression of the regulon, with maximum induction in the host cell cytosol and repression in the environmental habitat. Regulation operates through changes in PrfA activity-presumably by cofactor-mediated allosteric shift-and concentration, and involves transcriptional, translational and post-translational control mechanisms. There is evidence that PrfA exerts a more global influence on L. monocytogenes physiology via indirect mechanisms.


Assuntos
Listeria monocytogenes/genética , Listeriose/microbiologia , Fatores de Terminação de Peptídeos/genética , Sequência de Aminoácidos , Animais , Humanos , Listeria monocytogenes/patogenicidade , Modelos Moleculares , Dados de Sequência Molecular , Fatores de Terminação de Peptídeos/química , Fatores de Terminação de Peptídeos/fisiologia , Regulon/genética , Virulência/genética
12.
Mol Microbiol ; 59(2): 415-32, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16390439

RESUMO

Listeria ivanovii differs from the human pathogen Listeria monocytogenes in that it specifically affects ruminants, causing septicaemia and abortion but not meningo-encephalitis. The genetic characterization of spontaneous L. ivanovii mutants lacking the virulence factor SmcL (sphingomyelinase) led us to identify LIPI-2, the first species-specific pathogenicity island from Listeria. Besides SmcL, this 22 kb chromosomal locus encodes 10 internalin (Inl) proteins: i-InlB1 and -B2 are large/surface-associated Inls similar to L. monocytogenes InlB; i-InlE to -L are small/excreted (SE)-Inls, i-InlG being a tandem fusion of two SE-Inls. Except i-inlB1, all LIPI-2 inl genes are controlled by the virulence regulator, PrfA. LIPI-2 is inserted into a tRNA locus and is unstable - half of it deleting at approximately 10(-4) frequency with a portion of contiguous DNA. The spontaneous mutants were attenuated in vivo in mice and lambs and showed impaired intracellular growth and apoptosis induction in bovine MDBK cells. Targeted knock-out mutations associated the virulence defect with LIPI-2 genes. The region between the core genome loci ysnB-tRNA(arg) and ydeI flanking LIPI-2 contained different gene complements in the different Listeria spp. and even serovars of L. monocytogenes, including remnants of the PSA bacteriophage int gene in serovar 4b, indicating it is a hot spot for horizontal genome diversification. LIPI-2 is conserved in L. ivanovii ssp. ivanovii and londoniensis, suggesting an early acquisition during the species' evolution. LIPI-2 is likely to play an important role in the pathogenic and host tropism of L. ivanovii.


Assuntos
Proteínas de Bactérias/genética , Deleção de Genes , Genes Bacterianos , Genoma Bacteriano , Listeria/genética , Esfingomielina Fosfodiesterase/genética , Sequência de Bases , Mapeamento Cromossômico , Listeria/patogenicidade , Virulência/genética
13.
Appl Environ Microbiol ; 71(12): 9008-12, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16332910

RESUMO

We describe a novel quantitative real-time (Q)-PCR assay for Listeria monocytogenes based on the coamplification of a target hly gene fragment and an internal amplification control (IAC). The IAC is a chimeric double-stranded DNA containing a fragment of the rapeseed BnACCg8 gene flanked by the hly-specific target sequences. This IAC is detected using a second TaqMan probe labeled with a different fluorophore, enabling the simultaneous monitoring of the hly and IAC signals. The hly-IAC assay had a specificity and sensitivity of 100%, as assessed using 49 L. monocytogenes isolates of different serotypes and 96 strains of nontarget bacteria, including 51 Listeria isolates. The detection and quantification limits were 8 and 30 genome equivalents, and the coefficients for PCR linearity (R2) and efficiency (E) were 0.997 and 0.80, respectively. We tested the performance of the hly-IAC Q-PCR assay using various broth media and food matrices. Fraser and half-Fraser media, raw pork, and raw or cold-smoked salmon were strongly PCR-inhibitory. This Q-PCR assay for L. monocytogenes, the first incorporating an IAC to be described for quantitative detection of a food-borne pathogen, is a simple and robust tool facilitating the identification of false negatives or underestimations of contamination loads due to PCR failure.


Assuntos
Listeria monocytogenes/genética , Produtos da Carne/microbiologia , Reação em Cadeia da Polimerase/métodos , Animais , Toxinas Bacterianas/genética , Primers do DNA , Microbiologia de Alimentos , Amplificação de Genes , Proteínas de Choque Térmico/genética , Proteínas Hemolisinas , Suínos
14.
J Biol Chem ; 280(41): 35011-7, 2005 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-16093240

RESUMO

Sphingomyelinases C are enzymes that catalyze the hydrolysis of sphingomyelin in biological membranes to ceramide and phosphorylcholine. Various pathogenic bacteria produce secreted neutral sphingomyelinases C that act as membrane-damaging virulence factors. Mammalian neutral sphingomyelinases C, which display sequence homology to the bacterial enzymes, are involved in sphingolipid metabolism and signaling. This article describes the first structure to be determined for a member of the neutral sphingomyelinase C family, SmcL, from the intracellular bacterial pathogen Listeria ivanovii. The structure has been refined to 1.9-A resolution with phases derived by single isomorphous replacement with anomalous scattering techniques from a single iridium derivative. SmcL adopts a DNase I-like fold, and is the first member of this protein superfamily to have its structure determined that acts as a phospholipase. The structure reveals several unique features that adapt the protein to its phospholipid substrate. These include large hydrophobic beta-hairpin and hydrophobic loops surrounding the active site that may bind and penetrate the lipid bilayer to position sphingomyelin in a catalytically competent position. The structure also provides insight into the proposed general base/acid catalytic mechanism, in which His-325 and His-185 play key roles.


Assuntos
Listeria/enzimologia , Esfingomielina Fosfodiesterase/química , Sequência de Aminoácidos , Sítios de Ligação , Ceramidas/química , Clonagem Molecular , Cristalografia por Raios X , DNA/química , Elétrons , Hidrólise , Bicamadas Lipídicas/química , Modelos Moleculares , Dados de Sequência Molecular , Fosfolipases/química , Fosfolipídeos/química , Fosforilcolina/química , Reação em Cadeia da Polimerase , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Esfingomielinas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA