Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Mol Biol Cell ; 34(4): ar28, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36735487

ABSTRACT

The lymphatic circulation regulates transfer of tissue fluid and immune cells toward the venous circulation. While obesity impairs lymphatic vessel function, the contribution of lymphatic endothelial cells (LEC) to metabolic disease phenotypes is poorly understood. LEC of lymphatic microvessels are in direct contact with the interstitial fluid, whose composition changes during the development of obesity, markedly by increases in saturated fatty acids. Palmitate, the most prevalent saturated fatty acid in lymph and blood, is detrimental to metabolism and function of diverse tissues, but its impact on LEC function is relatively unknown. Here, palmitate (but not its unsaturated counterpart palmitoleate) destabilized adherens junctions in human microvascular LEC in culture, visualized as changes in VE-cadherin, α-catenin, and ß-catenin localization. Detachment of these proteins from cortical actin filaments was associated with abundant actomyosin stress fibers. The effects were Rho-associated protein kinase (ROCK)- and myosin-dependent, as inhibition with Y27632 or blebbistatin, respectively, prevented stress fiber accumulation and preserved junctions. Without functional junctions, palmitate-treated LEC failed to directionally migrate to close wounds in two dimensions and failed to form endothelial tubes in three dimensions. A reorganization of the lymphatic endothelial actin cytoskeleton may contribute to lymphatic dysfunction in obesity and could be considered as a therapeutic target.


Subject(s)
Endothelial Cells , Fatty Acids , Humans , Actin Cytoskeleton , Actomyosin , Adherens Junctions , Cadherins
2.
Mol Biol Cell ; 33(12): ar106, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35921166

ABSTRACT

Endothelia determine blood-to-tissue solute delivery, yet glucose transit is poorly understood. To illuminate mechanisms, we tracked [3H]-2-deoxyglucose (2-DG) in human adipose-tissue microvascular endothelial cells. 2-DG uptake was largely facilitated by the glucose transporters GLUT1 and GLUT3. Once in the cytosol, >80% of 2-DG became phosphorylated and ∼20% incorporated into glycogen, suggesting that transported glucose is readily accessible to cytosolic enzymes. Interestingly, a fraction of intracellular 2-DG was released over time (15-20% over 30 min) with slower kinetics than for uptake, involving GLUT3. In contrast to intracellular 2-DG, the released 2-DG was largely unphosphorylated. Glucose release involved endoplasmic reticulum-resident translocases/phosphatases and was stimulated by adrenaline, consistent with participation of glycogenolysis and glucose dephosphorylation. Surprisingly, the fluorescent glucose derivative 2-NBD-glucose (2-NBDG) entered cells largely via fluid phase endocytosis and exited by recycling. 2-NBDG uptake was insensitive to GLUT1/GLUT3 inhibition, suggesting poor influx across membranes. 2-NBDG recycling, but not 2-DG efflux, was sensitive to N-ethyl maleimide. In sum, by utilizing radioactive and fluorescent glucose derivatives, we identified two parallel routes of entry: uptake into the cytosol through dedicated glucose transporters and endocytosis. This reveals the complex glucose handling by endothelial cells that may contribute to glucose delivery to tissues.


Subject(s)
Deoxyglucose , Endothelial Cells , Deoxyglucose/pharmacology , Epinephrine , Glucose/pharmacology , Glucose Transporter Type 1 , Glucose Transporter Type 3 , Glycogen , Humans , Maleimides , Phosphoric Monoester Hydrolases
3.
Mol Microbiol ; 117(5): 1173-1195, 2022 05.
Article in English | MEDLINE | ID: mdl-35344242

ABSTRACT

Aluminum salts have been successfully utilized as adjuvants to enhance the immunogenicity of vaccine antigens since the 1930s. However, the cellular mechanisms behind the immune adjuvanticity effect of these materials in antigen-presenting cells are poorly understood. In this study, we investigated the uptake and trafficking of aluminum oxy-hydroxide (AlOOH), in RAW 264.7 murine and U-937 human macrophages-like cells. Furthermore, we determined the impact that the adsorption to AlOOH particulates has on the trafficking of a Bordetella pertussis vaccine candidate, the genetically detoxified pertussis toxin (gdPT). Our results indicate that macrophages internalize AlOOH by constitutive macropinocytosis assisted by the filopodial protrusions that capture the adjuvant particles. Moreover, we show that AlOOH has the capacity to nonspecifically adsorb IgG, engaging opsonic phagocytosis, which is a feature that may allow for more effective capture and uptake of adjuvant particles by antigen-presenting cells (APCs) at the site of vaccine administration. We found that AlOOH traffics to endolysosomal compartments that hold degradative properties. Importantly, while we show that gdPT escapes degradative endolysosomes and traffics toward the retrograde pathway, as reported for the wild-type pertussis toxin, the adsorption to AlOOH diverts gdPT to traffic to the adjuvant's lysosome-type compartments, which may be key for MHC-II-driven antigen presentation and activation of CD4+ T cell. Thus, our findings establish a direct link between antigen adsorption to AlOOH and the intracellular trafficking of antigens within antigen-presenting cells and bring to light a new potential mechanism for aluminum adjuvancy. Moreover, the in-vitro single-cell approach described herein provides a general framework and tools for understanding critical attributes of other vaccine formulations.


Subject(s)
Aluminum Hydroxide , Aluminum , Adjuvants, Immunologic/pharmacology , Aluminum/pharmacology , Aluminum Hydroxide/pharmacology , Animals , Humans , Lysosomes , Macrophages , Mice , Pertussis Toxin/genetics , Pertussis Toxin/pharmacology , Pertussis Vaccine/pharmacology
4.
Sci Rep ; 9(1): 13234, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31519919

ABSTRACT

Activated α2-macroglobulin (α2M*) and its receptor, low-density lipoprotein receptor-related protein 1 (LRP1), have been linked to proliferative retinal diseases. In Müller glial cells (MGCs), the α2M*/LRP1 interaction induces cell signaling, cell migration, and extracellular matrix remodeling, processes closely associated with proliferative disorders. However, the mechanism whereby α2M* and LRP1 participate in the aforementioned pathologies remains incompletely elucidated. Here, we investigate whether α2M* regulates both the intracellular distribution and sorting of LRP1 to the plasma membrane (PM) and how this regulation is involved in the cell migration of MGCs. Using a human Müller glial-derived cell line, MIO-M1, we demonstrate that the α2M*/LRP1 complex is internalized and rapidly reaches early endosomes. Afterward, α2M* is routed to degradative compartments, while LRP1 is accumulated at the PM through a Rab10-dependent exocytic pathway regulated by PI3K/Akt. Interestingly, Rab10 knockdown reduces both LRP1 accumulation at the PM and cell migration of MIO-M1 cells induced by α2M*. Given the importance of MGCs in the maintenance of retinal homeostasis, unravelling this molecular mechanism can potentially provide new therapeutic targets for the treatment of proliferative retinopathies.


Subject(s)
Cell Membrane/metabolism , Ependymoglial Cells/metabolism , Exocytosis , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , alpha-Macroglobulins/metabolism , rab GTP-Binding Proteins/metabolism , Cell Movement , Cells, Cultured , Ependymoglial Cells/cytology , Humans , Protein Transport , Signal Transduction
5.
J Biol Chem ; 294(44): 16172-16185, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31515271

ABSTRACT

Connections between deficient autophagy and insulin resistance have emerged, however, the mechanism through which reduced autophagy impairs insulin-signaling remains unknown. We examined mouse embryonic fibroblasts lacking Atg16l1 (ATG16L1 KO mouse embryonic fibroblasts (MEFs)), an essential autophagy gene, and observed deficient insulin and insulin-like growth factor-1 signaling. ATG16L1 KO MEFs displayed reduced protein content of insulin receptor substrate-1 (IRS1), pivotal to insulin signaling, whereas IRS1myc overexpression recovered downstream insulin signaling. Endogenous IRS1 protein content and insulin signaling were restored in ATG16L1 KO mouse embryonic fibroblasts (MEF) upon proteasome inhibition. Through proximity-dependent biotin identification (BioID) and co-immunoprecipitation, we found that Kelch-like proteins KLHL9 and KLHL13, which together form an E3 ubiquitin (Ub) ligase complex with cullin 3 (CUL3), are novel IRS1 interactors. Expression of Klhl9 and Klhl13 was elevated in ATG16L1 KO MEFs and siRNA-mediated knockdown of Klhl9, Klhl13, or Cul3 recovered IRS1 expression. Moreover, Klhl13 and Cul3 knockdown increased insulin signaling. Notably, adipose tissue of high-fat fed mice displayed lower Atg16l1 mRNA expression and IRS1 protein content, and adipose tissue KLHL13 and CUL3 expression positively correlated to body mass index in humans. We propose that ATG16L1 deficiency evokes insulin resistance through induction of Klhl9 and Klhl13, which, in complex with Cul3, promote proteasomal IRS1 degradation.


Subject(s)
Autophagy-Related Proteins/deficiency , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Animals , Autophagy/physiology , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Cullin Proteins/metabolism , Fibroblasts/metabolism , Genes, Regulator , HEK293 Cells , Humans , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Proteins/metabolism , Signal Transduction , Ubiquitin-Protein Ligase Complexes/metabolism
6.
Traffic ; 20(6): 390-403, 2019 06.
Article in English | MEDLINE | ID: mdl-30950163

ABSTRACT

An endothelial cell monolayer separates interstitia from blood and lymph, and determines the bidirectional transfer of solutes and macromolecules across these biological spaces. We review advances in transport modalities across these endothelial barriers. Glucose is a major fuel for the brain and peripheral tissues, and insulin acts on both central and peripheral tissues to promote whole-body metabolic signalling and anabolic activity. Blood-brain barrier endothelial cells display stringent tight junctions and lack pinocytic activity. Delivery of blood glucose and insulin to the brain occurs through their respective carrier (Glucose transporter 1) and receptor (insulin receptor), enacting bona fide transcytosis. At supraphysiological concentrations, insulin is also likely transferred by fluid phase cellular uptake and paracellular transport, especially in peripheral microvascular endothelia. The lymphatic microvasculature also transports insulin but in this case from tissues to lymph and therefrom to blood. This serves to end the hormone's action and to absorb highly concentrated subcutaneously injected insulin in diabetic individuals. The former function may involve receptor-mediated transcytosis into lymphatic endothelial cells, the latter fluid phase uptake and paracellular transport. Lymphatic capillaries also mediate carrier-dependent transport of other nutrients and macromolecules. These findings challenge the notion that lymphatic capillaries only transport macromolecules through intercellular flaps.


Subject(s)
Capillary Permeability , Endothelial Cells/metabolism , Insulin/metabolism , Transcytosis , Adipose Tissue/metabolism , Animals , Blood-Brain Barrier/cytology , Blood-Brain Barrier/metabolism , Humans , Insulin/blood , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism
7.
Am J Physiol Endocrinol Metab ; 315(2): E204-E217, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29509435

ABSTRACT

Whereas the blood microvasculature constitutes a biological barrier to the action of blood-borne insulin on target tissues, the lymphatic microvasculature might act as a barrier to subcutaneously administrated insulin reaching the circulation. Here, we evaluate the interaction of insulin with primary microvascular endothelial cells of lymphatic [human dermal lymphatic endothelial cells (HDLEC)] and blood [human adipose microvascular endothelial cells (HAMEC)] origin, derived from human dermal and adipose tissues, respectively. HDLEC express higher levels of insulin receptor and signal in response to insulin as low as 2.5 nM, while HAMEC only activate signaling at 100 nM (a dose that blood vessels do not normally encounter). Low insulin acts specifically through the insulin receptor, while supraphysiological insulin acts through both the IR and insulin growth factor-1 receptor. At supraphysiological or injection site-compatible doses pertinent to lymphatic microvessels, insulin enters HAMEC and HDLEC via fluid-phase endocytosis. Conversely, at physiologically circulating doses (0.2 nM) pertinent to blood microvessels, insulin enters HAMEC through a receptor-mediated process requiring IR autophosphorylation but not downstream insulin signaling. At physiological doses, internalized insulin is barely degraded and is instead released intact to the extracellular medium. In conclusion, we document for the first time the mechanism of interaction of insulin with lymphatic endothelial cells, which may be relevant to insulin absorption during therapeutic injections. Furthermore, we describe distinct action and uptake routes for insulin at physiological and supraphysiological doses in blood microvascular endothelial cells, providing a potential explanation for previously conflicting studies on endothelial insulin uptake.


Subject(s)
Blood Vessels/cytology , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Lymphatic Vessels/cytology , Microvessels/drug effects , Microvessels/metabolism , Adipose Tissue/blood supply , Adipose Tissue/cytology , Blood Vessels/metabolism , Cells, Cultured , Endocytosis/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Insulin/metabolism , Lymphatic Vessels/metabolism , Receptor, Insulin/metabolism , Skin/cytology , Skin/drug effects
8.
Methods Mol Biol ; 1713: 175-192, 2018.
Article in English | MEDLINE | ID: mdl-29218526

ABSTRACT

GLUT4 is the major glucose transporter in skeletal muscle. GLUT4 cycles to and from the plasma membrane and its exocytic rate is accelerated by insulin and muscle contraction to achieve a new steady state with more GLUT4 proteins at the muscle cell surface. To gain a better understanding of the molecular and cellular mechanisms that govern GLUT4 protein recycling, we developed an in vitro model in which myc-epitope-tagged GLUT4 or GLUT4-GFP is expressed in L6 skeletal muscle cells. The myc-epitope is inserted into an exofacial domain that is accessible to anti-myc antibodies from the outside of non-permeabilized cells, allowing one to count the number of transporters at the cell surface. This enables one to perform single-cell analysis using confocal fluorescence microscopy to quantify cell surface GLUT4myc or GLUT4myc-GFP in cells co-transfected with diverse cDNA constructs, treated with siRNAs, or co-stained with antibodies for other proteins of interest. Herein, we describe the methodology to perform these experimental approaches in insulin-stimulated L6 muscle cells.


Subject(s)
Glucose Transporter Type 4/metabolism , Muscle Fibers, Skeletal/metabolism , Single-Cell Analysis , Animals , Cell Line , Cell Membrane/metabolism , Epitopes/immunology , Epitopes/metabolism , Fluorescent Antibody Technique , Gene Expression , Genes, Reporter , Glucose/metabolism , Glucose Transporter Type 4/genetics , Molecular Imaging/methods , Protein Transport , RNA, Small Interfering/genetics , Rats , Single-Cell Analysis/methods , Transfection , Transport Vesicles/metabolism
9.
Trends Endocrinol Metab ; 28(8): 597-611, 2017 08.
Article in English | MEDLINE | ID: mdl-28602209

ABSTRACT

Glucose transport is rate limiting for dietary glucose utilization by muscle and fat. The glucose transporter GLUT4 is dynamically sorted and retained intracellularly and redistributes to the plasma membrane (PM) by insulin-regulated vesicular traffic, or 'GLUT4 translocation'. Here we emphasize recent findings in GLUT4 translocation research. The application of total internal reflection fluorescence microscopy (TIRFM) has increased our understanding of insulin-regulated events beneath the PM, such as vesicle tethering and membrane fusion. We describe recent findings on Akt-targeted Rab GTPase-activating proteins (GAPs) (TBC1D1, TBC1D4, TBC1D13) and downstream Rab GTPases (Rab8a, Rab10, Rab13, Rab14, and their effectors) along with the input of Rac1 and actin filaments, molecular motors [myosinVa (MyoVa), myosin1c (Myo1c), myosinIIA (MyoIIA)], and membrane fusion regulators (syntaxin4, munc18c, Doc2b). Collectively these findings reveal novel events in insulin-regulated GLUT4 traffic.


Subject(s)
Cytoplasmic Vesicles/metabolism , Glucose Transporter Type 4/metabolism , Insulin/physiology , Animals , Cytoplasmic Vesicles/drug effects , Humans , Insulin/pharmacology , Mice , Protein Transport/drug effects
10.
J Cell Biochem ; 118(7): 1810-1818, 2017 07.
Article in English | MEDLINE | ID: mdl-28012205

ABSTRACT

Distinct modes of cell migration contribute to diverse types of cell movements. The mesenchymal mode is characterized by a multistep cycle of membrane protrusion, the formation of focal adhesion, and the stabilization at the leading edge associated with the degradation of extracellular matrix (ECM) components and with regulated extracellular proteolysis. Both α2 -Macroglobulin (α2 M) and its receptor, low density lipoprotein receptor-related protein 1 (LRP1), play important roles in inflammatory processes, by controlling the extracellular activity of several proteases. The binding of the active form of α2 M (α2 M*) to LRP1 can also activate different signaling pathways in macrophages, thus inducing extracellular matrix metalloproteinase-9 (MMP-9) activation and cellular proliferation. In the present study, we investigated whether the α2 M*/LRP1 interaction induces cellular migration of the macrophage-derived cell line, Raw264.7. By using the wound-scratch migration assay and confocal microscopy, we demonstrate that α2 M* induces LRP1-mediated mesenchymal cellular migration. This migration exhibits the production of enlarged cellular protrusions, MT1-MMP distribution to these leading edge protrusions, actin polymerization, focal adhesion formation, and increased intracellular LRP1/ß1-integrin colocalization. Moreover, the presence of calphostin-C blocked the α2 M*-stimulated cellular protrusions, suggesting that the PKC activation is involved in the cellular motility of Raw264.7 cells. These findings could constitute a therapeutic target for inflammatory processes with deleterious consequences for human health, such as rheumatoid arthritis, atherosclerosis and cancer. J. Cell. Biochem. 118: 1810-1818, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Macrophages/cytology , Macrophages/drug effects , alpha-Macroglobulins/pharmacology , Animals , Cell Movement/drug effects , Cytoprotection/drug effects , Enzyme Inhibitors/pharmacology , Extracellular Matrix/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Microscopy, Confocal , Naphthalenes/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , RAW 264.7 Cells , Signal Transduction/drug effects
11.
Mol Biol Cell ; 27(1): 75-89, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26538022

ABSTRACT

Insulin promotes glucose uptake into skeletal muscle through recruitment of glucose transporter 4 (GLUT4) to the plasma membrane. Rab GTPases are molecular switches mobilizing intracellular vesicles, and Rab13 is necessary for insulin-regulated GLUT4-vesicle exocytic translocation in muscle cells. We show that Rab13 engages the scaffold protein MICAL-L2 in this process. RNA interference-mediated knockdown of MICAL-L2 or truncated MICAL-L2 (MICAL-L2-CT) impaired insulin-stimulated GLUT4 translocation. Insulin increased Rab13 binding to MICAL-L2, assessed by pull down and colocalization under confocal fluorescence and structured illumination microscopies. Association was also visualized at the cell periphery using TIRF microscopy. Insulin further increased binding of MICAL-L2 to α-actinin-4 (ACTN4), a protein involved in GLUT4 translocation. Rab13, MICAL-L2, and ACTN4 formed an insulin-dependent complex assessed by pull down and confocal fluorescence imaging. Of note, GLUT4 associated with the complex in response to insulin, requiring the ACTN4-binding domain in MICAL-L2. This was demonstrated by pull down with distinct fragments of MICAL-L2 and confocal and structured illumination microscopies. Finally, expression of MICAL-L2-CT abrogated the insulin-dependent colocalization of Rab13 with ACTN4 or Rab13 with GLUT4. Our findings suggest that MICAL-L2 is an effector of insulin-activated Rab13, which links to GLUT4 through ACTN4, localizing GLUT4 vesicles at the muscle cell periphery to enable their fusion with the membrane.


Subject(s)
Actinin/metabolism , GTP Phosphohydrolases/metabolism , Glucose Transporter Type 4/metabolism , Microfilament Proteins/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Exocytosis/physiology , Humans , Insulin/metabolism , Insulin/pharmacology , Muscle Cells/metabolism , Protein Binding , Protein Transport , Rats , Signal Transduction , Tight Junctions/metabolism , rab GTP-Binding Proteins/metabolism
12.
Invest Ophthalmol Vis Sci ; 56(11): 6948-60, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26513500

ABSTRACT

PURPOSE: In ischemic proliferative retinopathies, Müller glial cells (MGCs) acquire migratory abilities. However, the mechanisms that regulate this migration remain poorly understood. In addition, proliferative disorders associated with enhanced activities of matrix metalloproteinases (MMPs) also involve insulin-like growth factor (IGF)-1 participation. Therefore, the main interest of this work was to investigate the IGF-1 effect on the extracellular proteolytic activity in MGCs. METHODS: Cell culture supernatants and cell lysates of the human MGC line MIO-M1 stimulated with IGF-1 were analyzed for MMP-2 by zymographic and Western blot analysis. The MGCs' motility was evaluated by scratch wound assay. The MMP-2, ß1-integrin, and focal adhesions were detected by confocal microscopy. The localization of active MMPs and actin cytoskeleton were evaluated by in situ zymography. RESULTS: The IGF-1 induced the activation of canonical signaling pathways through the IGF-1R phosphorylation. Culture supernatants showed a relative decrease in the active form of MMP-2, correlating with an increased accumulation of MMP-2 protein in the MGCs' lysate. The IGF-1 effect on MMP-2 was abolished by an IGF-1R blocking antibody, αIR3, as well as by the PI3-kinase inhibitor, LY294002. The IGF-1 increased the migratory capacity of MGCs, which was blocked by the GM6001 MMP inhibitor, LY294002 and αIR3. Finally, IGF-1 induced the intracellular distribution of MMP-2 toward cellular protrusions and the partial colocalization with ß1-integrin and phospo-focal adhesion kinase signals. Gelatinase activity was concentrated along F-actin filaments. CONCLUSIONS: Taken together, these data indicate that IGF-1, through its receptor activation, regulates MGCs' motility by a mechanism that involves the MMP-2 and PI3K signaling pathway.


Subject(s)
Ependymoglial Cells/physiology , Insulin-Like Growth Factor I/physiology , Matrix Metalloproteinase 2/metabolism , Blotting, Western , Cell Line , Cell Movement/physiology , Enzyme Activation/physiology , Ependymoglial Cells/enzymology , Ependymoglial Cells/metabolism , Fluorescent Antibody Technique , Humans , Integrin beta1/physiology , Matrix Metalloproteinase 2/physiology , Microscopy, Confocal , Signal Transduction/physiology
13.
Cytometry A ; 85(7): 601-10, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24639232

ABSTRACT

In this article, we present a flow cytometry assay by which human blood monocyte subpopulations-classical (CD14(++) CD16(-)), intermediate (CD14(++) CD16(+)), and nonclassical (CD14(+) CD16(++)) monocytes-can be determined. Monocytic cells were selected from CD45(+) leukocyte subsets by differential staining of the low-density lipoprotein receptor-related protein 1 (LRP1), which allows reducing the spill-over of natural killer cells and granulocytes into the CD16(+) monocyte gate. Percentages of monocyte subpopulations established by this procedure were significantly comparable with those obtained by a well-standardized flow cytometry assay based on the HLA-DR monocyte-gating strategy. We also demonstrated that LRP1 is differentially expressed at cell surface of monocyte subpopulations, being significantly lower in nonclassical monocytes than in classical and intermediate monocytes. Cell surface expression of LRP1 accounts for only 20% of the total cellular content in each monocyte subpopulation. Finally, we established the within-individual biological variation (bCV%) of circulating monocyte subpopulations in healthy donors, obtaining values of 21%, 20%, and 17% for nonclassical, intermediate, and classical monocytes, respectively. Similar values of bCV% for LRP1 measured in each monocyte subpopulation were also obtained, suggesting that its variability is mainly influenced by the intrinsic biological variation of circulating monocytes. Thus, we conclude that LRP1 can be used as a third pan-monocytic marker together with CD14 and CD16 to properly identify monocyte subpopulations. The combined determination of monocyte subpopulations and LRP1 monocytic expression may be relevant for clinical studies of inflammatory processes, with special interest in atherosclerosis and cardiovascular disease.


Subject(s)
Flow Cytometry/methods , Low Density Lipoprotein Receptor-Related Protein-1/biosynthesis , Monocytes/classification , Monocytes/metabolism , Adult , Antibodies, Monoclonal , Atherosclerosis/diagnosis , Biomarkers/metabolism , Cardiovascular Diseases/diagnosis , Female , GPI-Linked Proteins/metabolism , Humans , Inflammation , Leukocyte Count , Leukocytes/metabolism , Lipopolysaccharide Receptors/metabolism , Male , Middle Aged , Monocytes/cytology , Receptors, IgG/metabolism , Young Adult
14.
FASEB J ; 27(8): 3181-97, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23640058

ABSTRACT

In retinal proliferative diseases, Müller glial cells (MGCs) acquire migratory abilities. However, the mechanisms that regulate this migration remain poorly understood. In addition, proliferative disorders associated with enhanced activities of matrix metalloprotease 2 (MMP-2) and MMP-9 also present increased levels of the protease inhibitor α2-macroglobulin (α2M) and its receptor, the low-density lipoprotein receptor-related protein 1 (LRP1). In the present work, we investigated whether the protease activated form of α2M, α2M*, and LRP1 are involved with the MGC migratory process. By performing wound-scratch migration and zymography assays, we demonstrated that α2M* induced cell migration and proMMP-2 activation in the human Müller glial cell line, MIO-M1. This induction was blocked when LRP1 and MT1-MMP were knocked down with siRNA techniques. Using fluorescence microscopy and biochemical procedures, we found that α2M* induced an increase in LRP1 and MT1-MMP accumulation in early endosomes, followed by endocytic recycling and intracellular distribution of MT1-MMP toward cellular protrusions. Moreover, Rab11-dominant negative mutant abrogated MT1-MMP recycling pathway, cell migration, and proMMP-2 activation induced by α2M*. In conclusion, α2M*, through its receptor LRP1, induces cellular migration of Müller glial cells by a mechanism that involves MT1-MMP intracellular traffic to the plasma membrane by a Rab11-dependent recycling pathway.


Subject(s)
Cell Movement/drug effects , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Matrix Metalloproteinase 14/metabolism , alpha-Macroglobulins/pharmacology , Carrier Proteins/pharmacology , Cell Line , Cell Membrane/metabolism , Endocytosis/drug effects , Endosomes/metabolism , Glutathione Transferase/pharmacology , Humans , Immunoblotting , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Matrix Metalloproteinase 14/genetics , Microscopy, Confocal , Models, Biological , Mutation , Neuroglia/cytology , Protein Binding/drug effects , Protein Transport/drug effects , RNA Interference , Recombinant Fusion Proteins/pharmacology , Signal Transduction/drug effects , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
15.
Acta bioquím. clín. latinoam ; 42(4): 529-538, oct.-dic. 2008. graf, tab
Article in Spanish | LILACS | ID: lil-633059

ABSTRACT

La determinación de Subpoblaciones Linfocitarias Humanas (SLH) por Citometría de Flujo, Linfocitos CD3+ (LT), LTCD4+, LTCD8+ y la relación LTCD4+/LTCD8+, permiten monitorear pacientes infectados con virus de la inmunodeficiencia humana (VIH), los cuales son comparados con Valores de Referencia (VR). A su vez los VR informados por la bibliografía son variables debido a la falta de validación analítica y partición estadística de los datos de VR, entre otros. En este trabajo, utilizando un ensayo validado (ISO15189), se establecieron VR para SLH con criterios estadísticos de partición en individuos con serología negativa para VIH (397 mujeres (F) y 279 varones (M), edad: 17-83 años). Las SLH fueron medidas en un Citómetro de Flujo (Coulter Epics XL-MCL). Los datos fueron procesados empleando como criterio estadístico de partición el algoritmo de Martin Gellerstedt (AMG). Del análisis estadístico y del AMG se determinó la partición de los datos en 6 subpoblaciones definidas por edad (intervalo de años) y género (F y M): grupos I y IV: 17-35 (F y M); grupos II y V: 36-50 (F y M); y grupos III y VI: >50 años (F y M). Para los 6 grupos se definieron VR de linfocitos totales, LT, LTCD4+, LTCD8+ y LTCD4+/LTCD8+. Para cada límite inferior y superior de VR se estableció el intervalo de confianza al 90%. En conclusión, este estudio permitió establecer VR para SLH en un ensayo validado empleando un criterio estadístico de partición, lo cual permitiría incrementar la confiabilidad de los resultados y uniformar a nivel internacional los VR en SLH en diferentes poblaciones.


The determination of Human Lymphocyte Subpopulations (HLS) including Lymphocytes CD3+ (LT), LTCD4+, LTCD8+ and LTCD4+/LTCD8+ ratio by flow cytometry, makes it possible to monitor patients infected who HIV, which are compared against reference values (RV). However, RV reported by the international bibliography are variable due to the absence of two main factors: analytical validation procedures and partitioning statistical criteria. This study, using a validated method (ISO15189), RV were established for HLS in individuals with negative serology for HIV (397 females (F) and 279 males (M), age: 17-83 years) applying partitioning statistical criteria. The values of HLS were obtained by flow cytometry (Coulter Epics XL-MCL). The data were processed using partitioning statistical criteria based on Martin Gellerstedt's algorithm (MGA). From the statistical analysis and MGA the partition of the data was determined in 6 subpopulations defined by age (interval of years) and gender (F and M): groups I and IV: 17-35 (F and M); groups II and V: 36-50 (F and M); and groups III and VI: >50 years (F and M). Hence, RV of total lymphocytes, LT, LTCD4+, LTCD8+ and LTCD4+/LTCD8+ ratio were defined. For each limit (lower and upper) of reference values, the 90% confidence interval was determined. In conclusion, this study allowed establishing RV for HLS in a validated method using a partitioning statistical criterion, which would allow increasing the assurance results and establishing an international criterion for reference values in HLS in different populations.


Subject(s)
Humans , Male , Female , Adolescent , Adult , Middle Aged , Aged , Aged, 80 and over , CD4-Positive T-Lymphocytes , Lymphocyte Subsets , CD4-CD8 Ratio/methods , Flow Cytometry/standards , Reference Values , Acquired Immunodeficiency Syndrome/immunology , Flow Cytometry/statistics & numerical data
16.
Cytometry B Clin Cytom ; 72(3): 223-6, 2007 May.
Article in English | MEDLINE | ID: mdl-17420983

ABSTRACT

BACKGROUND: In direct techniques of flow cytometry, the optimal antibody dilution or titer point is established from the plateau area of the antibody titration curve. However, the plateau area is defined without any statistical criteria, which may lead to an incorrect selection of antibody dilution. Herein, we report statistical criteria to establish the optimal antibody dilution for CD14, CD8, CD4, and CD3 analysis by flow cytometry in peripheral whole blood. METHODS: The unpaired t-test (two-tail P value) was used as statistical criteria to analyze the titration curve of the following monoclonal antibody panels: CD14-FITC, CD8-FITC, CD4-RD1, and CD3-PC5. RESULTS: Using the unpaired t-test (two-tail P value), the plateau area from the antibody titration curve was fitted when two consecutive antibody volumes showed mean peak of channel fluorescence (MPCF) values not significantly different. When the antibody was used at volume corresponding to that of the antibody titration point, the flow cytometry analysis of whole blood samples with different density of cell antigens can be correctly discriminated. CONCLUSION: This statistical criteria allows the fitting of the plateau area of MPCF versus antibody volume and consequently, to define the optimal antibody dilution.


Subject(s)
Antibodies/analysis , Data Interpretation, Statistical , Flow Cytometry/methods , Indicator Dilution Techniques , Titrimetry/methods , Antibodies/blood , Antibodies/chemistry , CD3 Complex/analysis , CD4 Antigens/analysis , CD8 Antigens/analysis , Humans , Lipopolysaccharide Receptors/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...