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.
Infect Immun ; 86(11)2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30104213

RESUMO

Listeria monocytogenes is a foodborne pathogen that infects the placenta and can cause pregnancy complications. Listeriosis usually occurs as a sporadic infection, but large outbreaks are also reported. Virulence from clinical isolates is rarely analyzed due to the large number of animals required, but this knowledge could help guide the response to an outbreak. We implemented a DNA barcode system using signature tags that allowed us to efficiently assay variations in virulence across a large number of isolates. We tested 77 signature-tagged clones of clinical L. monocytogenes strains from 72 infected human placentas and 5 immunocompromised patients, all of which were isolated since 2000. These strains were tested for virulence in a modified competition assay in comparison to that of the laboratory strain 10403S. We used two in vivo models of listeriosis: the nonpregnant mouse and the pregnant guinea pig. Strains that were frequently found at a high abundance within infected organs were considered hypervirulent, while strains frequently found at a low abundance were considered hypovirulent. Virulence split relatively evenly among hypovirulent strains, hypervirulent strains, and strains as virulent as 10403S. The laboratory strain was found to have an intermediate virulence phenotype, supporting its suitability for use in pathogenesis studies. Further, we found that splenic virulence and placental virulence are closely linked in both the guinea pig and mouse models. This suggests that outbreak and sporadic pregnancy-associated L. monocytogenes strains are not generally more virulent than lab reference strains. However, some strains did show consistent and reproducible virulence differences, suggesting that their further study may reveal deeper insights into the biological underpinnings of listeriosis.


Assuntos
Listeria monocytogenes/patogenicidade , Listeriose/microbiologia , Listeriose/patologia , Complicações Infecciosas na Gravidez/microbiologia , Complicações Infecciosas na Gravidez/patologia , Fatores de Virulência/análise , Estruturas Animais/microbiologia , Animais , Modelos Animais de Doenças , Feminino , Cobaias , Humanos , Listeria monocytogenes/genética , Listeria monocytogenes/crescimento & desenvolvimento , Listeria monocytogenes/isolamento & purificação , Camundongos , Placenta/microbiologia , Gravidez , Virulência , Fatores de Virulência/genética
2.
PLoS Pathog ; 14(5): e1007094, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29847585

RESUMO

During pregnancy, the placenta protects the fetus against the maternal immune response, as well as bacterial and viral pathogens. Bacterial pathogens that have evolved specific mechanisms of breaching this barrier, such as Listeria monocytogenes, present a unique opportunity for learning how the placenta carries out its protective function. We previously identified the L. monocytogenes protein Internalin P (InlP) as a secreted virulence factor critical for placental infection. Here, we show that InlP, but not the highly similar L. monocytogenes internalin Lmo2027, binds to human afadin (encoded by AF-6), a protein associated with cell-cell junctions. A crystal structure of InlP reveals several unique features, including an extended leucine-rich repeat (LRR) domain with a distinctive Ca2+-binding site. Despite afadin's involvement in the formation of cell-cell junctions, MDCK epithelial cells expressing InlP displayed a decrease in the magnitude of the traction stresses they could exert on deformable substrates, similar to the decrease in traction exhibited by AF-6 knock-out MDCK cells. L. monocytogenes ΔinlP mutants were deficient in their ability to form actin-rich protrusions from the basal face of polarized epithelial monolayers, a necessary step in the crossing of such monolayers (transcytosis). A similar phenotype was observed for bacteria expressing an internal in-frame deletion in inlP (inlP ΔLRR5) that specifically disrupts its interaction with afadin. However, afadin deletion in the host cells did not rescue the transcytosis defect. We conclude that secreted InlP targets cytosolic afadin to specifically promote L. monocytogenes transcytosis across the basal face of epithelial monolayers, which may contribute to the crossing of the basement membrane during placental infection.


Assuntos
Proteínas de Bactérias/metabolismo , Membrana Basal/microbiologia , Listeria monocytogenes/patogenicidade , Proteínas dos Microfilamentos/metabolismo , Complicações Infecciosas na Gravidez/metabolismo , Animais , Feminino , Feto/microbiologia , Humanos , Listeriose/metabolismo , Proteínas de Membrana/metabolismo , Placenta/metabolismo , Placenta/microbiologia , Gravidez , Fatores de Virulência/metabolismo
3.
Infect Immun ; 86(6)2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29483290

RESUMO

Intrauterine infections lead to serious complications for mother and fetus, including preterm birth, maternal and fetal death, and neurological sequelae in the surviving offspring. Improving maternal and child heath is a global priority. Yet, the development of strategies to prevent and treat pregnancy-related diseases has lagged behind progress made in other medical fields. One of the challenges is finding tractable model systems that replicate the human maternal-fetal interface. Animal models offer the ability to study pathogenesis and host defenses in vivo However, the anatomy of the maternal-fetal interface is highly divergent across species. While many tools are available to study host responses in the pregnant mouse model, other animals have placentas that are more similar to that of humans. Here we describe new developments in animal and human tissue models to investigate the pathogenesis of listeriosis at the maternal-fetal interface. We highlight gaps in existing knowledge and make recommendations on how they can be filled.


Assuntos
Listeria monocytogenes/fisiologia , Listeriose/transmissão , Modelos Biológicos , Complicações Infecciosas na Gravidez/microbiologia , Técnicas de Cultura de Tecidos , Animais , Feminino , Humanos , Listeriose/microbiologia , Troca Materno-Fetal , Placenta/microbiologia , Gravidez
4.
PLoS One ; 9(4): e91711, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24743728

RESUMO

Informed behavior change as an HIV prevention tool has yielded unequal successes across populations. Despite decades of HIV education, some individuals remain at high risk. The mainstream media often portrays these risk factors as products of race and national borders; however, a rich body of recent literature proposes a host of complex social factors that influence behavior, including, but not limited to: poverty, income inequality, stigmatizing social institutions and health care access. We examined the relationship between numerous social indicators and HIV incidence across eighty large U.S. cities in 1990 and 2000. During this time, major correlating factors included income inequality, poverty, educational attainment, residential segregation and marriage rates. However, these ecological factors were weighted differentially across risk groups (e.g. heterosexual, intravenous drug use, men who have sex with men (MSM)). Heterosexual risk rose significantly with poor economic indicators, while MSM risk depended more heavily on anti-homosexual stigma (as measured by same-sex marriage laws). HIV incidence among black individuals correlated significantly with numerous economic factors but also with segregation and imbalances in the male:female ratio (often an effect of mass incarceration). Our results support an overall model of HIV ecology where poverty, income inequality and social inequality (in the form of institutionalized racism and anti-homosexual stigma) have over time developed into synergistic drivers of disease transmission in the U.S., inhibiting information-based prevention efforts. The relative weights of these distal factors vary over time and by HIV risk group. Our testable model may be more generally applicable within the U.S. and beyond.


Assuntos
Infecções por HIV/epidemiologia , Fatores Sociológicos , Escolaridade , Feminino , Heterossexualidade/estatística & dados numéricos , Homossexualidade Masculina/estatística & dados numéricos , Humanos , Renda/estatística & dados numéricos , Masculino , Casamento/estatística & dados numéricos , Pobreza/estatística & dados numéricos , Características de Residência/estatística & dados numéricos , Assunção de Riscos , Abuso de Substâncias por Via Intravenosa/virologia , Estados Unidos/epidemiologia
5.
PLoS Pathog ; 9(12): e1003821, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24348256

RESUMO

Fetal syncytiotrophoblasts form a unique fused multinuclear surface that is bathed in maternal blood, and constitutes the main interface between fetus and mother. Syncytiotrophoblasts are exposed to pathogens circulating in maternal blood, and appear to have unique resistance mechanisms against microbial invasion. These are due in part to the lack of intercellular junctions and their receptors, the Achilles heel of polarized mononuclear epithelia. However, the syncytium is immune to receptor-independent invasion as well, suggesting additional general defense mechanisms against infection. The difficulty of maintaining and manipulating primary human syncytiotrophoblasts in culture makes it challenging to investigate the cellular and molecular basis of host defenses in this unique tissue. Here we present a novel system to study placental pathogenesis using murine trophoblast stem cells (mTSC) that can be differentiated into syncytiotrophoblasts and recapitulate human placental syncytium. Consistent with previous results in primary human organ cultures, murine syncytiotrophoblasts were found to be resistant to infection with Listeria monocytogenes via direct invasion and cell-to-cell spread. Atomic force microscopy of murine syncytiotrophoblasts demonstrated that these cells have a greater elastic modulus than mononuclear trophoblasts. Disruption of the unusually dense actin structure--a diffuse meshwork of microfilaments--with Cytochalasin D led to a decrease in its elastic modulus by 25%. This correlated with a small but significant increase in invasion of L. monocytogenes into murine and human syncytium. These results suggest that the syncytial actin cytoskeleton may form a general barrier against pathogen entry in humans and mice. Moreover, murine TSCs are a genetically tractable model system for the investigation of specific pathways in syncytial host defenses.


Assuntos
Células Gigantes/microbiologia , Listeria monocytogenes/crescimento & desenvolvimento , Listeriose/imunologia , Placenta/citologia , Placenta/microbiologia , Complicações Infecciosas na Gravidez/imunologia , Animais , Fenômenos Biofísicos/imunologia , Células Cultivadas , Feminino , Células Gigantes/imunologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Transmissão Vertical de Doenças Infecciosas , Listeria monocytogenes/imunologia , Listeriose/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Placenta/imunologia , Gravidez , Complicações Infecciosas na Gravidez/microbiologia , Trofoblastos/citologia , Trofoblastos/imunologia , Trofoblastos/microbiologia , Células U937
6.
Curr Opin Microbiol ; 15(1): 36-43, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22169833

RESUMO

Placental infections are major causes of maternal and fetal disease. This review introduces a new paradigm for placental infections based on current knowledge of placental defenses and how this barrier can be breached. Transmission of pathogens from mother to fetus can occur at two sites of direct contact between maternal cells and specialized fetal cells (trophoblasts) in the human placenta: firstly, maternal immune and endothelial cells juxtaposed to extravillous trophoblasts in the uterine implantation site and secondly, maternal blood surrounding the syncytiotrophoblast (SYN). Recent findings suggest that the primary vulnerability is in the implantation site. We explore evidence that the placental SYN evolved as a defense against pathogens, and that inflammation-mediated spontaneous abortion may benefit mother and pathogen.


Assuntos
Infecções Bacterianas/imunologia , Placenta/imunologia , Complicações Infecciosas na Gravidez/imunologia , Complicações Infecciosas na Gravidez/microbiologia , Complicações Parasitárias na Gravidez/imunologia , Complicações Parasitárias na Gravidez/parasitologia , Infecções por Protozoários/imunologia , Aborto Séptico/imunologia , Aborto Séptico/microbiologia , Aborto Séptico/parasitologia , Infecções Bacterianas/microbiologia , Infecções Bacterianas/patologia , Feminino , Humanos , Placenta/microbiologia , Placenta/parasitologia , Gravidez , Complicações Infecciosas na Gravidez/parasitologia , Infecções por Protozoários/parasitologia , Infecções por Protozoários/patologia , Trofoblastos/imunologia
7.
Infect Immun ; 80(1): 418-28, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22083708

RESUMO

Toxoplasma gondii is a ubiquitous, obligate intracellular parasite capable of crossing the placenta to cause spontaneous abortion, preterm labor, or significant disease in the surviving neonate. Exploration of the cellular and histological components of the placental barrier is in its infancy, and both how and where T. gondii breaches it are unknown. The human placenta presents two anatomical interfaces between maternal cells and fetal cells (trophoblasts): (i) the villous region where maternal blood bathes syncytialized trophoblasts for nutrient exchange and (ii) the maternal decidua, where mononuclear, extravillous trophoblasts anchor the villous region to the uterus. Using first-trimester human placental explants, we demonstrate that the latter site is significantly more vulnerable to infection, despite presenting a vastly smaller surface. This is consistent with past findings concerning two vertically transmitted viruses and one bacterium. We further explore whether three genetically distinct T. gondii types (I, II, and III) are capable of preferential placental infection and survival in this model. We find no difference in these strains' ability to infect placental explants; however, slightly slower growth is evident in type II (Prugniaud [Pru]) parasites relative to other cell types, although this did not quite achieve statistical significance.


Assuntos
Placenta/imunologia , Toxoplasma/imunologia , Toxoplasma/patogenicidade , Toxoplasmose/imunologia , Feminino , Humanos , Modelos Biológicos , Técnicas de Cultura de Órgãos , Placenta/anatomia & histologia , Placenta/citologia , Gravidez
8.
PLoS Pathog ; 7(3): e1002005, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21408203

RESUMO

Listeria monocytogenes is a facultative intracellular bacterial pathogen that can infect the placenta, a chimeric organ made of maternal and fetal cells. Extravillous trophoblasts (EVT) are specialized fetal cells that invade the uterine implantation site, where they come into direct contact with maternal cells. We have shown previously that EVT are the preferred site of initial placental infection. In this report, we infected primary human EVT with L. monocytogenes. EVT eliminated ∼80% of intracellular bacteria over 24-hours. Bacteria were unable to escape into the cytoplasm and remained confined to vacuolar compartments that became acidified and co-localized with LAMP1, consistent with bacterial degradation in lysosomes. In human placental organ cultures bacterial vacuolar escape rates differed between specific trophoblast subpopulations. The most invasive EVT-those that would be in direct contact with maternal cells in vivo-had lower escape rates than trophoblasts that were surrounded by fetal cells and tissues. Our results suggest that EVT present a bottleneck in the spread of L. monocytogenes from mother to fetus by inhibiting vacuolar escape, and thus intracellular bacterial growth. However, if L. monocytogenes is able to spread beyond EVT it can find a more hospitable environment. Our results elucidate a novel aspect of the maternal-fetal barrier.


Assuntos
Listeria monocytogenes/crescimento & desenvolvimento , Listeria monocytogenes/patogenicidade , Placenta/citologia , Placenta/microbiologia , Trofoblastos/citologia , Trofoblastos/microbiologia , Células Cultivadas , Feminino , Feto/microbiologia , Imunofluorescência , Humanos , Transmissão Vertical de Doenças Infecciosas , Lisossomos/metabolismo , Microscopia Eletrônica de Transmissão , Gravidez , Técnicas de Cultura de Tecidos , Útero/microbiologia , Vacúolos/microbiologia
9.
PLoS Pathog ; 6(1): e1000732, 2010 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-20107601

RESUMO

Listeria monocytogenes is an important cause of maternal-fetal infections and serves as a model organism to study these important but poorly understood events. L. monocytogenes can infect non-phagocytic cells by two means: direct invasion and cell-to-cell spread. The relative contribution of each method to placental infection is controversial, as is the anatomical site of invasion. Here, we report for the first time the use of first trimester placental organ cultures to quantitatively analyze L. monocytogenes infection of the human placenta. Contrary to previous reports, we found that the syncytiotrophoblast, which constitutes most of the placental surface and is bathed in maternal blood, was highly resistant to L. monocytogenes infection by either internalin-mediated invasion or cell-to-cell spread. Instead, extravillous cytotrophoblasts-which anchor the placenta in the decidua (uterine lining) and abundantly express E-cadherin-served as the primary portal of entry for L. monocytogenes from both extracellular and intracellular compartments. Subsequent bacterial dissemination to the villous stroma, where fetal capillaries are found, was hampered by further cellular and histological barriers. Our study suggests the placenta has evolved multiple mechanisms to resist pathogen infection, especially from maternal blood. These findings provide a novel explanation why almost all placental pathogens have intracellular life cycles: they may need maternal cells to reach the decidua and infect the placenta.


Assuntos
Transmissão Vertical de Doenças Infecciosas , Listeriose/transmissão , Doenças Placentárias/microbiologia , Trofoblastos/microbiologia , Feminino , Imunofluorescência , Humanos , Processamento de Imagem Assistida por Computador , Listeria monocytogenes , Microscopia Confocal , Técnicas de Cultura de Órgãos , Gravidez , Complicações Infecciosas na Gravidez/microbiologia
10.
PLoS One ; 5(1): e8610, 2010 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-20062534

RESUMO

BACKGROUND: Recent studies have suggested that autophagy is utilized by cells as a protective mechanism against Listeria monocytogenes infection. METHODOLOGY/PRINCIPAL FINDINGS: However we find autophagy has no measurable role in vacuolar escape and intracellular growth in primary cultured bone marrow derived macrophages (BMDMs) deficient for autophagy (atg5-/-). Nevertheless, we provide evidence that the pore forming activity of the cholesterol-dependent cytolysin listeriolysin O (LLO) can induce autophagy subsequent to infection by L. monocytogenes. Infection of BMDMs with L. monocytogenes induced microtubule-associated protein light chain 3 (LC3) lipidation, consistent with autophagy activation, whereas a mutant lacking LLO did not. Infection of BMDMs that express LC3-GFP demonstrated that wild-type L. monocytogenes was encapsulated by LC3-GFP, consistent with autophagy activation, whereas a mutant lacking LLO was not. Bacillus subtilis expressing either LLO or a related cytolysin, perfringolysin O (PFO), induced LC3 colocalization and LC3 lipidation. Further, LLO-containing liposomes also recruited LC3-GFP, indicating that LLO was sufficient to induce targeted autophagy in the absence of infection. The role of autophagy had variable effects depending on the cell type assayed. In atg5-/- mouse embryonic fibroblasts, L. monocytogenes had a primary vacuole escape defect. However, the bacteria escaped and grew normally in atg5-/- BMDMs. CONCLUSIONS/SIGNIFICANCE: We propose that membrane damage, such as that caused by LLO, triggers bacterial-targeted autophagy, although autophagy does not affect the fate of wild-type intracellular L. monocytogenes in primary BMDMs.


Assuntos
Autofagia/fisiologia , Proteínas de Choque Térmico/fisiologia , Proteínas Hemolisinas/fisiologia , Listeriose/imunologia , Animais , Toxinas Bacterianas , Células Cultivadas , Lipossomos , Listeriose/fisiopatologia , Camundongos
11.
J Neurochem ; 94(5): 1288-96, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16000154

RESUMO

Transcription co-activators and histone acetyltransferases, p300 and cyclic AMP responsive element-binding protein-binding protein (CBP), participate in hypoxic activation of hypoxia-inducible genes. Here, we show that exposure of PC12 and cells to 1-10% oxygen results in hyperphosphorylation of p300/CBP. This response is fast, long lasting and specific for hypoxia, but not for hypoxia-mimicking agents such as desferioxamine or Co2+ ions. It is also cell-type specific and occurs in pheochromocytoma PC12 cells and the carotid body of rats but not in hepatoblastoma cells. The p300 hyperphosphorylation specifically depends on the release of intracellular calcium from inositol 1,4,5-triphosphate (IP3)-sensitive stores. However, it is not inhibited by pharmacological inhibitors of any of the kinases traditionally known to be directly or indirectly calcium regulated. On the other hand, p300 hyperphosphorylation is inhibited by several different inhibitors of the glucose metabolic pathway from generation of NADH by glyceraldehyde 3-phosphate dehydrogenase, through the transfer of NADH through the glycerol phosphate shuttle to ubiquinone and complex III of the mitochondrial respiratory chain. Inhibition of IP3-sensitive calcium stores decreases generation of ATP, and this inhibition is significantly stronger in hypoxia than in normoxia. We propose that the NADH glycerol phosphate shuttle participates in generating a pool of ATP that serves either as a co-factor or a modulator of the kinases involved in the phosphorylation of p300/CBP during hypoxia.


Assuntos
Hipóxia/metabolismo , Proteínas Nucleares/metabolismo , Transativadores/metabolismo , Animais , Cálcio/metabolismo , Corpo Carotídeo/efeitos dos fármacos , Corpo Carotídeo/metabolismo , Corpo Carotídeo/patologia , Linhagem Celular , Proteína p300 Associada a E1A , Glucose/metabolismo , Humanos , Hipóxia/patologia , Hipóxia/fisiopatologia , Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Oxigênio/farmacologia , Células PC12/efeitos dos fármacos , Células PC12/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley
12.
J Physiol ; 564(Pt 1): 131-43, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15677684

RESUMO

T lymphocytes are exposed to hypoxia during their development and when they migrate to hypoxic pathological sites. Although it has been shown that hypoxia inhibits Kv1.3 channels and proliferation in human T cells, the mechanisms by which hypoxia regulates T cell activation are not fully understood. Herein we test the hypothesis that hypoxic inhibition of Kv1.3 channels induces membrane depolarization, thus modulating the increase in cytoplasmic Ca2+ that occurs during activation. Hypoxia causes membrane depolarization in human CD3+ T cells, as measured by fluorescence-activated cell sorting (FACS) with the voltage-sensitive dye DiBAC4(3). Similar depolarization is produced by the selective Kv1.3 channel blockers ShK-Dap22 and margatoxin. Furthermore, pre-exposure to such blockers prevents any further depolarization by hypoxia. Since membrane depolarization is unfavourable to the influx of Ca2+ through the CRAC channels (necessary to drive many events in T cell activation such as cytokine production and proliferation), the effect of hypoxia on T cell receptor-mediated increase in cytoplasmic Ca2+ was determined using fura-2. Hypoxia depresses the increase in Ca2+ induced by anti-CD3/CD28 antibodies in approximately 50% of lymphocytes. In the remaining cells, hypoxia either did not elicit any change or produced a small increase in cytoplasmic Ca2+. Similar effects were observed in resting and pre-activated CD3+ cells and were mimicked by ShK-Dap22. These effects appear to be mediated solely by Kv1.3 channels, as we find no influence of hypoxia on IKCa1 and CRAC channels. Our findings indicate that hypoxia modulates Ca2+ homeostasis in T cells via Kv1.3 channel inhibition and membrane depolarization.


Assuntos
Ativação Linfocitária/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Receptores de Antígenos de Linfócitos T/fisiologia , Linfócitos T/metabolismo , Hipóxia Celular/fisiologia , Humanos , Canal de Potássio Kv1.3 , Potenciais da Membrana/fisiologia , Transdução de Sinais/imunologia , Linfócitos T/fisiologia , Fatores de Tempo
13.
Mol Biol Cell ; 15(5): 2312-23, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15004224

RESUMO

Using a biochemically complex cytoplasmic extract to reconstitute actin-based motility of Listeria monocytogenes and polystyrene beads coated with the bacterial protein ActA, we have systematically varied a series of biophysical parameters and examined their effects on initiation of motility, particle speed, speed variability, and path trajectory. Bead size had a profound effect on all aspects of motility, with increasing size causing slower, straighter movement and inhibiting symmetry-breaking. Speed also was reduced by extract dilution, by addition of methylcellulose, and paradoxically by addition of excess skeletal muscle actin, but it was enhanced by addition of nonmuscle (platelet) actin. Large, persistent individual variations in speed were observed for all conditions and their relative magnitude increased with extract dilution, indicating that persistent alterations in particle surface properties may be responsible for intrinsic speed variations. Trajectory curvature was increased for smaller beads and also for particles moving in the presence of methylcellulose or excess skeletal muscle actin. Symmetry breaking and movement initiation occurred by two distinct modes: either stochastic amplification of local variation for small beads in concentrated extracts, or gradual accumulation of strain in the actin gel for large beads in dilute extracts. Neither mode was sufficient to enable spherical particles to break symmetry in the cytoplasm of living cells.


Assuntos
Actinas/fisiologia , Proteínas de Bactérias/química , Listeria monocytogenes/fisiologia , Proteínas de Membrana/química , Actinas/química , Animais , Proteínas de Bactérias/fisiologia , Toxinas Bacterianas/química , Bioensaio , Fenômenos Biofísicos , Biofísica , Extratos Celulares/química , Linhagem Celular , Cães , Proteínas de Fluorescência Verde/análise , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/fisiologia , Proteínas Hemolisinas , Listeria monocytogenes/química , Proteínas de Membrana/fisiologia , Microesferas , Movimento , Músculo Esquelético/química , Poliestirenos/química , Viscosidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...