Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Biomed Opt ; 28(6): 066502, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37351197

RESUMO

Significance: Fluorescence lifetime imaging microscopy (FLIM) of the metabolic co-enzyme nicotinamide adenine dinucleotide (phosphate) [NAD(P)H] is a popular method to monitor single-cell metabolism within unperturbed, living 3D systems. However, FLIM of NAD(P)H has not been performed in a light-sheet geometry, which is advantageous for rapid imaging of cells within live 3D samples. Aim: We aim to design, validate, and demonstrate a proof-of-concept light-sheet system for NAD(P)H FLIM. Approach: A single-photon avalanche diode camera was integrated into a light-sheet microscope to achieve optical sectioning and limit out-of-focus contributions for NAD(P)H FLIM of single cells. Results: An NAD(P)H light-sheet FLIM system was built and validated with fluorescence lifetime standards and with time-course imaging of metabolic perturbations in pancreas cancer cells with 10 s integration times. NAD(P)H light-sheet FLIM in vivo was demonstrated with live neutrophil imaging in a larval zebrafish tail wound also with 10 s integration times. Finally, the theoretical and practical imaging speeds for NAD(P)H FLIM were compared across laser scanning and light-sheet geometries, indicating a 30× to 6× acquisition speed advantage for the light sheet compared to the laser scanning geometry. Conclusions: FLIM of NAD(P)H is feasible in a light-sheet geometry and is attractive for 3D live cell imaging applications, such as monitoring immune cell metabolism and migration within an organism.


Assuntos
NAD , Neoplasias Pancreáticas , Animais , NAD/metabolismo , Peixe-Zebra , Microscopia de Fluorescência/métodos , Fótons , Imagem Óptica/métodos
2.
bioRxiv ; 2023 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-36778488

RESUMO

Single photon avalanche diode (SPAD) array sensors can increase the imaging speed for fluorescence lifetime imaging microscopy (FLIM) by transitioning from laser scanning to widefield geometries. While a SPAD camera in epi-fluorescence geometry enables widefield FLIM of fluorescently labeled samples, label-free imaging of single-cell autofluorescence is not feasible in an epi-fluorescence geometry because background fluorescence from out-of-focus features masks weak cell autofluorescence and biases lifetime measurements. Here, we address this problem by integrating the SPAD camera in a light sheet illumination geometry to achieve optical sectioning and limit out-of-focus contributions, enabling fast label-free FLIM of single-cell NAD(P)H autofluorescence. The feasibility of this NAD(P)H light sheet FLIM system was confirmed with time-course imaging of metabolic perturbations in pancreas cancer cells with 10 s integration times, and in vivo NAD(P)H light sheet FLIM was demonstrated with live neutrophil imaging in a zebrafish tail wound, also with 10 s integration times. Finally, the theoretical and practical imaging speeds for NAD(P)H FLIM were compared across laser scanning and light sheet geometries, indicating a 30X to 6X frame rate advantage for the light sheet compared to the laser scanning geometry. This light sheet system provides faster frame rates for 3D NAD(P)H FLIM for live cell imaging applications such as monitoring single cell metabolism and immune cell migration throughout an entire living organism.

3.
Elife ; 112022 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-35200139

RESUMO

The function of macrophages in vitro is linked to their metabolic rewiring. However, macrophage metabolism remains poorly characterized in situ. Here, we used two-photon intensity and lifetime imaging of autofluorescent metabolic coenzymes, nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) and flavin adenine dinucleotide (FAD), to assess the metabolism of macrophages in the wound microenvironment. Inhibiting glycolysis reduced NAD(P)H mean lifetime and made the intracellular redox state of macrophages more oxidized, as indicated by reduced optical redox ratio. We found that TNFα+ macrophages had lower NAD(P)H mean lifetime and were more oxidized compared to TNFα- macrophages. Both infection and thermal injury induced a macrophage population with a more oxidized redox state in wounded tissues. Kinetic analysis detected temporal changes in the optical redox ratio during tissue repair, revealing a shift toward a more reduced redox state over time. Metformin reduced TNFα+ wound macrophages, made intracellular redox state more reduced and improved tissue repair. By contrast, depletion of STAT6 increased TNFα+ wound macrophages, made redox state more oxidized and impaired regeneration. Our findings suggest that autofluorescence of NAD(P)H and FAD is sensitive to dynamic changes in intracellular metabolism in tissues and can be used to probe the temporal and spatial regulation of macrophage metabolism during tissue damage and repair.


Assuntos
Flavina-Adenina Dinucleotídeo/metabolismo , Macrófagos/metabolismo , NADP/metabolismo , Ferimentos e Lesões/metabolismo , Peixe-Zebra/metabolismo , Animais , Feminino , Fluorescência , Glicólise , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Oxirredução , Fator de Necrose Tumoral alfa/metabolismo
4.
J Cell Biol ; 220(8)2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34047769

RESUMO

Neutrophil recruitment to tissue damage is essential for host defense but can also impede tissue repair. The cues that differentially regulate neutrophil responses to tissue damage and infection remain unclear. Here, we report that the paracrine factor myeloid-derived growth factor (MYDGF) is induced by tissue damage and regulates neutrophil motility to damaged, but not infected, tissues in zebrafish larvae. Depletion of MYDGF impairs wound healing, and this phenotype is rescued by depleting neutrophils. Live imaging and photoconversion reveal impaired neutrophil reverse migration and inflammation resolution in mydgf mutants. We found that persistent neutrophil inflammation in tissues of mydgf mutants was dependent on the HIF-1α pathway. Taken together, our data suggest that MYDGF is a damage signal that regulates neutrophil interstitial motility and inflammation through a HIF-1α pathway in response to tissue damage.


Assuntos
Nadadeiras de Animais/metabolismo , Movimento Celular , Inflamação/metabolismo , Interleucinas/metabolismo , Infiltração de Neutrófilos , Neutrófilos/metabolismo , Cicatrização , Infecção dos Ferimentos/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Nadadeiras de Animais/lesões , Nadadeiras de Animais/microbiologia , Nadadeiras de Animais/patologia , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/genética , Inflamação/microbiologia , Interleucinas/genética , Macrófagos/metabolismo , Macrófagos/microbiologia , Microscopia de Fluorescência , Neutrófilos/microbiologia , Comunicação Parácrina , Pseudomonas aeruginosa/patogenicidade , Transdução de Sinais , Fatores de Tempo , Infecção dos Ferimentos/genética , Infecção dos Ferimentos/microbiologia , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
5.
Cell Commun Signal ; 18(1): 144, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32900380

RESUMO

BACKGROUND: Lung cancer is the second most commonly occurring cancer. The ability to metastasize and spread to distant locations renders the tumor more aggressive. Members of the Rho subfamily of small GTP-binding proteins (GTPases) play a central role in the regulation of the actin cytoskeleton and in cancer cell migration and metastasis. In this study we investigated the role of the RhoA/Cdc42 GAP, StarD13, a previously described tumor suppressor, in malignancy, migration and invasion of the lung cancer cells A549. METHODS: We knocked down StarD13 expression in A549 lung cancer cells and tested the effect on cell migration and invadopodia formation using time lapse imaging and invasion assays. We also performed rescue experiments to determine the signaling pathways downstream of StarD13 and transfected the cells with FRET biosensors for RhoGTPases to identify the proteins involved in invadopodia formation. RESULTS: We observed a decrease in the level of expression of StarD13 in lung tumor tissues compared to normal lung tissues through immunohistochemistry. StarD13 also showed a lower expression in the lung adenocarcinoma cell line A549 compared to normal lung cells, WI38. In addition, the depletion of StarD13 increased cell proliferation and viability in WI38 and A549 cells, suggesting that StarD13 might potentially be a tumor suppressor in lung cancer. The depletion of StarD13, however, inhibited cell motility, conversely demonstrating a positive regulatory role in cell migration. This was potentially due to the constitutive activation of RhoA detected by pull down and FRET assays. Surprisingly, StarD13 suppressed cell invasion by inhibiting Cdc42-mediated invadopodia formation. Indeed, TKS4 staining and invadopodia assay revealed that StarD13 depletion increased Cdc42 activation as well as invadopodia formation and matrix degradation. Normal lung cells depleted of StarD13 also produced invadopodia, otherwise a unique hallmark of invasive cancer cells. Cdc42 knock down mimicked the effects of StarD13, while overexpression of a constitutively active Cdc42 mimicked the effects of its depletion. Finally, immunostaining and FRET analysis revealed the absence of StarD13 in invadopodia as compared to Cdc42, which was activated in invadopodia at the sites of matrix degradation. CONCLUSION: In conclusion, StarD13 plays distinct roles in lung cancer cell migration and invasion through its differential regulation of Rho GTPases. Video abstract.


Assuntos
Adenocarcinoma de Pulmão/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Neoplasias Pulmonares/metabolismo , Podossomos/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Células A549 , Adenocarcinoma de Pulmão/patologia , Movimento Celular , Humanos , Neoplasias Pulmonares/patologia , Invasividade Neoplásica/patologia , Podossomos/patologia
6.
Methods Mol Biol ; 2108: 281-293, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31939189

RESUMO

Genetically encoded optogenetic tools are increasingly popular and useful for perturbing signaling pathways with high spatial and temporal resolution in living cells. Here, we show basic procedures employed to implement optogenetics of Rho GTPases in a macrophage cell line. Methods described here are generally applicable to other genetically encoded optogenetic tools utilizing the blue-green spectrum of light for activation, designed for specific proteins and enzymatic targets important for immune cell functions.


Assuntos
Luz , Macrófagos/metabolismo , Macrófagos/efeitos da radiação , Optogenética , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , Expressão Gênica , Genes Reporter , Camundongos , Microscopia de Fluorescência , Optogenética/métodos , Ligação Proteica , Células RAW 264.7 , Transfecção
7.
J Hepatol ; 70(4): 710-721, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30572006

RESUMO

BACKGROUND & AIMS: Non-alcoholic fatty liver disease/non-alcoholic steatohepatitis (NAFLD/NASH) is an increasing clinical problem associated with progression to hepatocellular carcinoma (HCC). The effect of a high-fat diet on the early immune response in HCC is poorly understood, while the role of metformin in treating NAFLD and HCC remains controversial. Herein, we visualized the early immune responses in the liver and the effect of metformin on progression of HCC using optically transparent zebrafish. METHODS: We used live imaging to visualize liver inflammation and disease progression in a NAFLD/NASH-HCC zebrafish model. We combined a high-fat diet with a transgenic zebrafish HCC model induced by hepatocyte-specific activated beta-catenin and assessed liver size, angiogenesis, micronuclei formation and inflammation in the liver. In addition, we probed the effects of metformin on immune cell composition and early HCC progression. RESULTS: We found that a high-fat diet induced an increase in liver size, enhanced angiogenesis, micronuclei formation and neutrophil infiltration in the liver. Although macrophage number was not affected by diet, a high-fat diet induced changes in macrophage morphology and polarization with an increase in liver associated TNFα-positive macrophages. Treatment with metformin altered macrophage polarization, reduced liver size and reduced micronuclei formation in NAFLD/NASH-associated HCC larvae. Moreover, a high-fat diet reduced T cell density in the liver, which was reversed by treatment with metformin. CONCLUSIONS: These findings suggest that diet alters macrophage polarization and exacerbates the liver inflammatory microenvironment and cancer progression in a zebrafish model of NAFLD/NASH-associated HCC. Metformin specifically affects the progression induced by diet and modulates the immune response by affecting macrophage polarization and T cell infiltration, suggesting possible effects of metformin on tumor surveillance. LAY SUMMARY: This paper reports a new zebrafish model that can be used to study the effects of diet on liver cancer. We found that a high-fat diet promotes non-resolving inflammation in the liver and enhances cancer progression. In addition, we found that metformin, a drug used to treat diabetes, inhibits high-fat diet-induced cancer progression in this model, by reducing diet-induced non-resolving inflammation and potentially restoring tumor surveillance.


Assuntos
Carcinoma Hepatocelular/complicações , Carcinoma Hepatocelular/tratamento farmacológico , Progressão da Doença , Imunidade Inata/efeitos dos fármacos , Neoplasias Hepáticas/complicações , Neoplasias Hepáticas/tratamento farmacológico , Metformina/uso terapêutico , Hepatopatia Gordurosa não Alcoólica/complicações , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Animais , Animais Geneticamente Modificados , Polaridade Celular/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Inflamação/tratamento farmacológico , Inflamação/etiologia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Metformina/farmacologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Peixe-Zebra
8.
Sci Rep ; 7(1): 8547, 2017 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-28819224

RESUMO

Macrophage interactions with other cells, either locally or at distances, are imperative in both normal and pathological conditions. While soluble means of communication can transmit signals between different cells, it does not account for all long distance macrophage interactions. Recently described tunneling nanotubes (TNTs) are membranous channels that connect cells together and allow for transfer of signals, vesicles, and organelles. However, very little is known about the mechanism by which these structures are formed. Here we investigated the signaling pathways involved in TNT formation by macrophages using multiple imaging techniques including super-resolution microscopy (3D-SIM) and live-cell imaging including the use of FRET-based Rho GTPase biosensors. We found that formation of TNTs required the activity and differential localization of Cdc42 and Rac1. The downstream Rho GTPase effectors mediating actin polymerization through Arp2/3 nucleation, Wiskott-Aldrich syndrome protein (WASP) and WASP family verprolin-homologous 2 (WAVE2) proteins are also important, and both pathways act together during TNT biogenesis. Finally, TNT function as measured by transfer of cellular material between cells was reduced following depletion of a single factor demonstrating the importance of these factors in TNTs. Given that the characterization of TNT formation is still unclear in the field; this study provides new insights and would enhance the understanding of TNT formation towards investigating new markers.


Assuntos
Actinas/metabolismo , Extensões da Superfície Celular/metabolismo , Macrófagos/metabolismo , Polimerização , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Comunicação Celular , Linhagem Celular , Humanos , Macrófagos/citologia , Camundongos , Transdução de Sinais , Imagem com Lapso de Tempo/métodos , Proteína da Síndrome de Wiskott-Aldrich/genética , Proteína da Síndrome de Wiskott-Aldrich/metabolismo , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
9.
Methods Mol Biol ; 1519: 125-143, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27815877

RESUMO

The p21-family members of Rho GTPases are important for the control of actin cytoskeleton dynamics, and are critical regulators of phagocytosis. The three-dimensional structure of phagosomes and the highly compartmentalized nature of the signaling mechanisms during phagocytosis require high-resolution imaging using ratiometric biosensors to decipher Rho GTPase activities regulating phagosome formation and function. Here we describe methods for the expression and ratiometric imaging of FRET-based Rho GTPase biosensors in macrophages during phagocytosis. As an example, we show Cdc42 activity at the phagosome over Z-serial planes. In addition, we demonstrate the usage of a new, fast, and user-friendly deconvolution package that delivers significant improvements in the attainable details of Rho GTPase activity in phagosome structures.


Assuntos
Técnicas Biossensoriais/métodos , Transferência Ressonante de Energia de Fluorescência/métodos , Fagocitose , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Ativação Enzimática , Imageamento Tridimensional , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Células RAW 264.7 , Estatística como Assunto , Proteína cdc42 de Ligação ao GTP/metabolismo
10.
Nature ; 539(7630): 575-578, 2016 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-27828948

RESUMO

Mitochondrial products such as ATP, reactive oxygen species, and aspartate are key regulators of cellular metabolism and growth. Abnormal mitochondrial function compromises integrated growth-related processes such as development and tissue repair, as well as homeostatic mechanisms that counteract ageing and neurodegeneration, cardiovascular disease, and cancer. Physiologic mechanisms that control mitochondrial activity in such settings remain incompletely understood. Here we show that the atypical Fat1 cadherin acts as a molecular 'brake' on mitochondrial respiration that regulates vascular smooth muscle cell (SMC) proliferation after arterial injury. Fragments of Fat1 accumulate in SMC mitochondria, and the Fat1 intracellular domain interacts with multiple mitochondrial proteins, including critical factors associated with the inner mitochondrial membrane. SMCs lacking Fat1 (Fat1KO) grow faster, consume more oxygen for ATP production, and contain more aspartate. Notably, expression in Fat1KO cells of a modified Fat1 intracellular domain that localizes exclusively to mitochondria largely normalizes oxygen consumption, and the growth advantage of these cells can be suppressed by inhibition of mitochondrial respiration, which suggest that a Fat1-mediated growth control mechanism is intrinsic to mitochondria. Consistent with this idea, Fat1 species associate with multiple respiratory complexes, and Fat1 deletion both increases the activity of complexes I and II and promotes the formation of complex-I-containing supercomplexes. In vivo, Fat1 is expressed in injured human and mouse arteries, and inactivation of SMC Fat1 in mice potentiates the response to vascular damage, with markedly increased medial hyperplasia and neointimal growth, and evidence of higher SMC mitochondrial respiration. These studies suggest that Fat1 controls mitochondrial activity to restrain cell growth during the reparative, proliferative state induced by vascular injury. Given recent reports linking Fat1 to cancer, abnormal kidney and muscle development, and neuropsychiatric disease, this Fat1 function may have importance in other settings of altered cell growth and metabolism.


Assuntos
Artérias/citologia , Artérias/metabolismo , Caderinas/metabolismo , Respiração Celular , Mitocôndrias/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Aorta/citologia , Aorta/lesões , Aorta/metabolismo , Artérias/lesões , Ácido Aspártico/metabolismo , Caderinas/química , Caderinas/deficiência , Proliferação de Células , Técnicas de Inativação de Genes , Humanos , Masculino , Camundongos , Mitocôndrias/química , Membranas Mitocondriais/metabolismo , Proteínas Mitocondriais/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/lesões , Músculo Liso Vascular/metabolismo , Neointima/metabolismo , Oxigênio/metabolismo , Consumo de Oxigênio
11.
J Immunol ; 196(8): 3479-93, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26951800

RESUMO

Despite the 92% homology of the hematopoietic cell-specific Rac2 to the canonical isoform Rac1, these isoforms have been shown to play nonredundant roles in immune cells. To study isoform-specific dynamics of Rac in live cells, we developed a genetically encoded, single-chain FRET-based biosensor for Rac2. We also made significant improvements to our existing single-chain Rac1 biosensor. We optimized the biosensor constructs for facile expression in hematopoietic cells and performed functional validations in murine macrophage sublines of RAW264.7 cells. Rac2, Rac1, and Cdc42 have been implicated in the formation of actin-rich protrusions by macrophages, but their individual activation dynamics have not been previously characterized. We found that both Rac1 and Rac2 had similar activation kinetics, yet they had distinct spatial distributions in response to the exogenous stimulus, fMLF. Active Rac1 was mainly localized to the cell periphery, whereas active Rac2 was distributed throughout the cell, with an apparent higher concentration in the perinuclear region. We also performed an extensive morphodynamic analysis of Rac1, Rac2, and Cdc42 activities during the extension of random protrusions. We found that Rac2 appears to play a leading role in the generation of random protrusions, as we observed an initial strong activation of Rac2 in regions distal from the leading edge, followed by the activation of Rac1, a second burst of Rac2 and then Cdc42 immediately behind the leading edge. Overall, isoform-specific biosensors that have been optimized for expression should be valuable for interrogating the coordination of Rho family GTPase activities in living cells.


Assuntos
Técnicas Biossensoriais/métodos , Transferência Ressonante de Energia de Fluorescência/métodos , Neuropeptídeos/genética , Isoformas de Proteínas/genética , Proteína cdc42 de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/genética , Animais , Linhagem Celular , Extensões da Superfície Celular/fisiologia , Células HEK293 , Humanos , Macrófagos/imunologia , Camundongos , Interferência de RNA , RNA Interferente Pequeno/genética , Proteína RAC2 de Ligação ao GTP
12.
J Cell Biol ; 205(5): 737-51, 2014 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-24891603

RESUMO

Invadopodia are actin-rich protrusions that degrade the extracellular matrix and are required for stromal invasion, intravasation, and metastasis. The role of the focal adhesion protein talin in regulating these structures is not known. Here, we demonstrate that talin is required for invadopodial matrix degradation and three-dimensional extracellular matrix invasion in metastatic breast cancer cells. The sodium/hydrogen exchanger 1 (NHE-1) is linked to the cytoskeleton by ezrin/radixin/moesin family proteins and is known to regulate invadopodium-mediated matrix degradation. We show that the talin C terminus binds directly to the moesin band 4.1 ERM (FERM) domain to recruit a moesin-NHE-1 complex to invadopodia. Silencing talin resulted in a decrease in cytosolic pH at invadopodia and blocked cofilin-dependent actin polymerization, leading to impaired invadopodium stability and matrix degradation. Furthermore, talin is required for mammary tumor cell motility, intravasation, and spontaneous lung metastasis in vivo. Thus, our findings provide a novel understanding of how intracellular pH is regulated and a molecular mechanism by which talin enhances tumor cell invasion and metastasis.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Proteínas dos Microfilamentos/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Talina/metabolismo , Actinas/metabolismo , Animais , Sítios de Ligação , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto , Matriz Extracelular/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Concentração de Íons de Hidrogênio , Neoplasias Pulmonares/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos SCID , Metástase Neoplásica , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo , Trocador 1 de Sódio-Hidrogênio
13.
Methods Mol Biol ; 1172: 173-84, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24908304

RESUMO

Cytokine stimulations of leukocytes many times result in transient activation of the p21 Rho family of small GTPases. The role of these molecules during cell migration and chemotaxis is well established. The traditional approach to study the activation dynamics of these proteins involves affinity pull-downs that are often cumbersome and prone to errors. Here, we describe a reagent and a method of simple "mix-and-measure" approach useful for determining the activation status of endogenous Cdc42 GTPase from cell lysates.


Assuntos
Bioensaio , Quimiocina CX3CL1/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Coloração e Rotulagem/métodos , Proteína cdc42 de Ligação ao GTP/genética , Animais , Extratos Celulares/química , Linhagem Celular , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Camundongos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo
14.
Methods Mol Biol ; 1172: 263-70, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24908313

RESUMO

Immunofluorescence is an important technique required to observe expression, localization and colocalization of proteins within the cell. Here we describe the immunofluorescence and subsequent confocal microscopy technique of tumor necrosis factor-α (TNF) in human neutrophils (polymorphonuclear leukocytes; PMN). The qualitative technique can be used to observe the expression pattern changes from resting to stimulated leukocytes. Colocalization with other cytokines, proteins, or organelles can be observed. This immunofluorescence technique can be done in 1-2 days.


Assuntos
Citoplasma/química , Neutrófilos/efeitos dos fármacos , Fator de Necrose Tumoral alfa/análise , Anticorpos/química , Núcleo Celular/química , Núcleo Celular/imunologia , Células Cultivadas , Citoplasma/imunologia , Imunofluorescência , Humanos , Lipopolissacarídeos/farmacologia , Microscopia Confocal , Neutrófilos/citologia , Neutrófilos/imunologia , Fixação de Tecidos , Fator de Necrose Tumoral alfa/biossíntese
15.
Nat Cell Biol ; 16(6): 574-86, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24859002

RESUMO

Rho family GTPases control cell migration and participate in the regulation of cancer metastasis. Invadopodia, associated with invasive tumour cells, are crucial for cellular invasion and metastasis. To study Rac1 GTPase in invadopodia dynamics, we developed a genetically encoded, single-chain Rac1 fluorescence resonance energy (FRET) transfer biosensor. The biosensor shows Rac1 activity exclusion from the core of invadopodia, and higher activity when invadopodia disappear, suggesting that reduced Rac1 activity is necessary for their stability, and Rac1 activation is involved in disassembly. Photoactivating Rac1 at invadopodia confirmed this previously unknown Rac1 function. We describe here an invadopodia disassembly model, where a signalling axis involving TrioGEF, Rac1, Pak1, and phosphorylation of cortactin, causes invadopodia dissolution. This mechanism is critical for the proper turnover of invasive structures during tumour cell invasion, where a balance of proteolytic activity and locomotory protrusions must be carefully coordinated to achieve a maximally invasive phenotype.


Assuntos
Neoplasias da Mama/enzimologia , Movimento Celular , Extensões da Superfície Celular/enzimologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Quinases Ativadas por p21/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Técnicas Biossensoriais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Extensões da Superfície Celular/patologia , Cortactina/metabolismo , Matriz Extracelular/metabolismo , Feminino , Transferência Ressonante de Energia de Fluorescência , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Invasividade Neoplásica , Proteínas do Tecido Nervoso/genética , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA , Ratos , Fatores de Tempo , Transfecção , Quinases Ativadas por p21/genética , Proteínas rac1 de Ligação ao GTP/genética
16.
Immunol Rev ; 256(1): 222-39, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24117824

RESUMO

Macrophages are best known for their protective search and destroy functions against invading microorganisms. These processes are commonly known as chemotaxis and phagocytosis. Both of these processes require actin cytoskeletal remodeling to produce distinct F-actin-rich membrane structures called lamellipodia and phagocytic cups. This review will focus on the mechanisms by which macrophages regulate actin polymerization through initial receptor signaling and subsequent Arp2/3 activation by nucleation-promoting factors like the WASP/WAVE family, followed by remodeling of actin networks to produce these very distinct structures.


Assuntos
Membrana Celular/metabolismo , Quimiotaxia/fisiologia , Macrófagos/fisiologia , Fagocitose/fisiologia , Citoesqueleto de Actina/fisiologia , Animais , Humanos , Antígeno de Macrófago 1/imunologia , Antígeno de Macrófago 1/metabolismo , Receptores de IgG/metabolismo
17.
Am J Transl Res ; 5(1): 103-15, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23390570

RESUMO

Bronchopulmonary dysplasia (BPD) is one of the most common causes of mortality and morbidity in neonatal intensive care units. Persistent inflammation, with an abnormal influx of polymorphonuclear leukocytes (PMNs) followed by monocytes (MONOs), occurs early in the pathogenesis of BPD. Anti-inflammatory therapy with better efficacy and safety than dexamethasone (DEX) is needed. In the present study we determined cell-specific gene expression and cytokine release in response to glucocorticoids versus interleukin-10 (IL-10). Subsequently, we hypothesized that the insensitivity of MONOs to DEX was associated with a failure of the glucocorticoid receptor to translocate to the nucleus. PMNs and MONOs were isolated from umbilical cord blood at birth, and pretreated with PBS vehicle, IL-10 or glucocorticoids prior to endotoxin (LPS)-stimulation for 4 and 18h. Genome-wide gene expressions were determined by microarray and validated by RT-qPCR. Interleukin 8 release in cell culture supernatant was measured by ELISA. To examine the mechanism of monocyte insensitivity to glucocorticoids, nuclear translocation of the glucocorticoid receptor was determined by Western blots. MONOs had 6 times the number of genes changing expression with IL-10 compared to PMNs at 4h. DEX at the therapeutic level for neonates with BPD had no effect on gene expression in MONOs. The order of potency for inhibition of interleukin-8 release from MONOs was IL-10 >betamethasone >dexamethasone and hydrocortisone. Glucocorticoid potency in MONOs was directly related to glucocorticoid receptor translocation to nucleus. Gene expression profiling for IL-10 versus glucocorticoids indicates there may be major differences in therapeutic efficacy for BPD.

18.
PLoS One ; 8(1): e53641, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23326478

RESUMO

INTRODUCTION: Increasing evidence now supports the association between the fetal inflammatory response syndrome (FIRS) with the pathogenesis of preterm labor, intraventricular hemorrhage and bronchopulmonary dysplasia. Polymorphonuclear leukocyte (PMNs) and mononuclear cell (MONOs) infiltration of the placenta is associated with these disorders. The aim of this study was to reveal cell-specific differences in gene expression and cytokine release in response to endotoxin that would elucidate inflammatory control mechanisms in the newly born. METHODS: PMNs and MONOs were separately isolated from the same cord blood sample. A genome-wide microarray screened for gene expression and related pathways at 4 h of LPS stimulation (n = 5). RT-qPCR and ELISA were performed for selected cytokines at 4 h and 18 h of LPS stimulation. RESULTS: Compared to PMNs, MONOs had a greater diversity and more robust gene expression that included pro-inflammatory (PI) cytokines, chemokines and growth factors at 4 h. Only MONOs had genes changing expression (all up regulated including interleukin-10) that were clustered in the JAK/STAT pathway. Pre-incubation with IL-10 antibody, for LPS-stimulated MONOs, led to up regulated PI and IL-10 gene expression and release of PI cytokines after 4 h. DISCUSSION: The present study suggests a dominant role of MONO gene expression in control of the fetal inflammatory response syndrome at 4 hrs of LPS stimulation. LPS-stimulated MONOs but not PMNs of the newborn have the ability to inhibit PI cytokine gene expression by latent IL-10 release.


Assuntos
Citocinas/genética , Endotoxinas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-10/metabolismo , Leucócitos/metabolismo , Nascimento a Termo/genética , Transcriptoma , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Genoma Humano/genética , Humanos , Recém-Nascido , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Leucócitos/efeitos dos fármacos , Monócitos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
19.
Neonatology ; 97(2): 108-16, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19713717

RESUMO

BACKGROUND: Monocytes play an important role in the fetal and neonatal inflammatory response syndrome. They are also the precursors of alveolar macrophages, microglial and Kupffer cells. Monocytes have pro-inflammatory (PI) and anti-inflammatory (AI) functions. Interleukin (IL)-10 is a potent AI cytokine released by monocytes. OBJECTIVE: We determined the effects of endogenous and exogenous IL-10 versus equimolar levels of dexamethasone (DEX) on PI and AI cytokine release, as well as transcription factor DNA-binding activity, in endotoxin (lipopolysaccharide, LPS)-stimulated monocytes of the newborn. METHODS: Monocytes were isolated into culture media from cord blood. ELISAs, electrophoretic mobility shift assays and Western blots were employed. RESULTS: LPS-stimulated monocyte release of PI cytokines, tumor necrosis factor-alpha (TNF-alpha), IL-1beta and IL-8, over 18 h was significantly augmented by addition of an IL-10 monoclonal antibody. Exogenous IL-10 at 10(-8)M inhibited PI cytokine release by 89-97%, while DEX at an equimolar level had no effect. DNA-binding activities of the PI transcription factors nuclear factor-kappaB (NF-kappaB) and activator protein-1 (AP-1), and the AI transcription factor signal transducer and activator of transcription 3 (STAT3) were induced over 18 h. DEX at 10(-8)M had no effect on any transcription factor DNA binding, but exogenous IL-10 at 10(-8)M produced a 60% inhibition of AP-1 DNA binding and enhanced phosphorylation of nuclear STAT3 for 18 h. CONCLUSION: At therapeutic levels of DEX, monocyte release of PI cytokine was insensitive to DEX in comparison to IL-10. IL-10 or its mechanism of action could lead to new therapy for inflammatory disorders in the perinatal period.


Assuntos
Dexametasona/farmacologia , Glucocorticoides/farmacologia , Interleucina-10/metabolismo , Interleucina-10/farmacologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Anticorpos Monoclonais/farmacologia , Western Blotting , Células Cultivadas , Relação Dose-Resposta a Droga , Ensaio de Desvio de Mobilidade Eletroforética , Ensaio de Imunoadsorção Enzimática , Sangue Fetal/citologia , Humanos , Recém-Nascido , Interleucina-10/imunologia , Interleucina-1beta/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos Alveolares/citologia , Monócitos/citologia , Fator de Transcrição STAT3/metabolismo , Fatores de Transcrição/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
20.
Pediatr Res ; 65(4): 425-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19127214

RESUMO

Interleukin-10 (IL-10), an anti-inflammatory cytokine, may have therapeutic potential in the fetal inflammatory response syndrome and its sequelae such as bronchopulmonary dysplasia (BPD). Our aim was to compare the effects of IL-10 versus dexamethasone (DEX) on important PMN functions of the newborn. PMNs were isolated into culture medium from cord blood after elective cesarean section deliveries. IL-10 and DEX were compared on an equimolar basis corresponding to previously measured plasma levels of DEX from infants treated for BPD. The endotoxin (LPS)-stimulated release of the pro-inflammatory cytokines, tumor necrosis factor (TNFalpha) and IL-1 beta, were markedly inhibited equally by IL-10 and DEX; the anti-inflammatory cytokine IL-4 was not released and IL-1 receptor antagonist (IL-1ra) was released less with DEX compared with IL-10. PMNs exposed to LPS, N-formyl-L-methionyl-L-leucyl-L-phenylalanine (fMLP), or S. aureus did not show a significant difference between control, DEX and IL-10 for apoptosis, respiratory burst, phagocytosis or killing respectively. Chemotaxis to fMLP or IL-8 was unaffected by DEX or IL-10. The principal effects of both IL-10 and DEX, on the PMN functions studied, are related to the control of pro- and anti-inflammatory cytokine release.


Assuntos
Anti-Inflamatórios/farmacologia , Dexametasona/farmacologia , Interleucina-10/farmacologia , Neutrófilos/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Células Cultivadas , Quimiotaxia de Leucócito/efeitos dos fármacos , Citocinas/metabolismo , Citotoxicidade Imunológica/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sangue Fetal/citologia , Humanos , Recém-Nascido , Lipopolissacarídeos/farmacologia , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Infiltração de Neutrófilos/efeitos dos fármacos , Neutrófilos/enzimologia , Neutrófilos/imunologia , Fagocitose/efeitos dos fármacos , Explosão Respiratória/efeitos dos fármacos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA