Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters











Publication year range
1.
Commun Biol ; 5(1): 748, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35902770

ABSTRACT

Allergies have become a rising health problem, where plentiful substances can trigger IgE-mediated allergies in humans. While profilins are considered minor allergens, these ubiquitous proteins are primary molecules involved in cross-reactivity and pollen-food allergy syndrome. Here we report the first crystal structures of murine Fab/IgE, with its chains naturally paired, in complex with the allergen profilin from Hevea brasiliensis (Hev b 8). The crystallographic models revealed that the IgE's six complementarity-determining regions (CDRs) interact with the allergen, comprising a rigid paratope-epitope surface of 926 Å2, which includes an extensive network of interactions. Interestingly, we also observed previously unreported flexibility at Fab/IgE's elbow angle, which did not influence the shape of the paratope. The Fab/IgE exhibits a high affinity for Hev b 8, even when using 1 M NaCl in BLI experiments. Finally, based on the encouraging cross-reactivity assays using two mutants of the maize profilin (Zea m 12), this antibody could be a promising tool in IgE engineering for diagnosis and research applications.


Subject(s)
Food Hypersensitivity , Profilins , Allergens/chemistry , Allergens/metabolism , Amino Acid Sequence , Animals , Contractile Proteins/metabolism , Humans , Immunoglobulin E , Mice , Microfilament Proteins/metabolism , Profilins/genetics , Profilins/metabolism
2.
Mini Rev Med Chem ; 22(18): 2419-2428, 2022.
Article in English | MEDLINE | ID: mdl-35264090

ABSTRACT

Interleukin-6 (IL-6) influences both inflammatory response and anti-inflammatory processes. This cytokine can be released by exercising skeletal muscle, which characterizes it as a myokine. Unlike what is observed in inflammation, IL-6 produced by skeletal muscle is not preceded by the release of other pro-inflammatory cytokines, but it seems to be dependent on the lactate produced during exercise, thus causing different effects from those seen in inflammatory state. After binding to its receptor, myokine IL-6 activates the PI3K-Akt pathway. One consequence of this upregulation is the potentiation of insulin signaling, which enhances insulin sensitivity. IL-6 increases GLUT-4 vesicle mobilization to the muscle cell periphery, increasing the glucose transport into the cell, and also glycogen synthesis. Muscle glycogen provides energy for ATP resynthesis, and regulates Ca2+ release by the sarcoplasmic reticulum, influencing muscle contraction, and, hence, muscle function by multiple pathways. Another implication for the upregulation of the PI3K-Akt pathway is the activation of mTORC1, which regulates mRNA translational efficiency by regulating translation machinery, and translational capacity by inducing ribosomal biogenesis. Thus, IL-6 may contribute to skeletal muscle hypertrophy and function by increasing contractile protein synthesis.


Subject(s)
Insulin Resistance , Interleukin-6 , Muscle, Skeletal , Adenosine Triphosphate/metabolism , Anti-Inflammatory Agents/pharmacology , Calcium/metabolism , Contractile Proteins/metabolism , Cytokines/metabolism , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Glycogen/metabolism , Humans , Hypertrophy/metabolism , Insulin/metabolism , Insulin Resistance/physiology , Interleukin-6/metabolism , Lactates/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism
3.
Clin Transl Oncol ; 22(4): 522-531, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31190277

ABSTRACT

BACKGROUND: The tumor microenvironment (TME) regulates tumor progression, and cancer-associated fibroblasts (CAFs) are the primary stromal components of the TME, with the potential to drive tumor metastasis via the secretion of paracrine factors, but the specific mechanisms driving this process have not been defined. METHODS: Proteins secreted from CAFs and normal fibroblasts (NFs) were analyzed via proteomic analysis (fold change > 2, p < 0.05) to identify tumor-promoting proteins secreted by CAFs. RESULTS: Proteomic analysis revealed that microfibrillar-associated protein 5 (MFAP5) is preferentially expressed and secreted by CAFs relative to NFs, which was confirmed by Western blotting and RT-qPCR. Transwell and wound healing assays confirmed that MFAP5 is secreted by CAFs, and drives the invasion and migration of MCF7 breast cancer cells. We further found that in MCF7 cells MFAP5 promoted epithelial-mesenchymal transition, activating Notch1 signaling and consequently upregulating NICD1 and slug. When Notch1 was knocked down in MCF7 cells, the ability of MFAP5 to promote invasion and migration decreased. CONCLUSION: CAFs promote cancer cells invasion and migration via MFAP5 secretion and activation of the Notch1/slug signaling. These data highlight this pathway as a therapeutic target to disrupt tumor progression through the interference of CAF-tumor crosstalk.


Subject(s)
Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/physiology , Contractile Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Receptor, Notch1/physiology , Snail Family Transcription Factors/physiology , Cell Movement , Epithelial-Mesenchymal Transition , Female , Humans , MCF-7 Cells , Neoplasm Invasiveness , Signal Transduction/physiology , Tumor Microenvironment
4.
Eur Spine J ; 28(6): 1413-1422, 2019 06.
Article in English | MEDLINE | ID: mdl-31069526

ABSTRACT

PURPOSE: The objective of this study was to analyze the layers of yellow ligament in lumbar canal stenosis and disk herniation. METHODS: Eighteen ligaments were harvested from patients with lumbar spinal canal stenosis. Twenty-nine normal samples from lumbar spine disk herniation patients served as control. All surgical procedures were the same. Ligaments were stained in hematoxylin and eosin; picrosirius-hematoxylin for collagen; Weigert's resorcin-fuchsin for elaunin, oxytalan and elastic fibers; and transmission electron microscopy. Immunohistochemistry was performed for Il-6; Il-10; and CD-31, PGP9.5. Results are described in means and standard error (mean ± SE), and all analyses adopted the significance level of P < 0.05. RESULTS: Spinal stenosis ligaments were 2.5 × thicker. Control superficial ligaments presented a large number of thick, compact collagen fibers and a significant amount of oxytalan and mature elastic fibers. The deep layer presented a large number of mature elastic fibers. In the stenosis group, collagen was thinner and compacted in both layers. There was no difference in the interleukin profile among groups. The deep portion of the stenosis group presented a higher number of vessels and nerves. CONCLUSION: Two layers compose the elastic system of the normal ligamentum flavum, where the deep portion is mainly responsible for its elasticity (elaunin fibers), while its resistance depends on the concentration of oxytalan fibers, which are more present in the superficial layer. Ligamentum flavum in the stenosis samples presents more mononuclear infiltrate and more degraded elastic fibers with a higher number of vessels in its deep portion. These slides can be retrieved under Electronic Supplementary Material.


Subject(s)
Intervertebral Disc Degeneration/metabolism , Ligamentum Flavum/chemistry , Lumbar Vertebrae/chemistry , Spinal Stenosis/metabolism , Adult , Aged , Aged, 80 and over , Contractile Proteins/analysis , Elastic Tissue/chemistry , Elastic Tissue/pathology , Elastic Tissue/ultrastructure , Elasticity , Extracellular Matrix Proteins/analysis , Female , Humans , Intervertebral Disc Degeneration/pathology , Intervertebral Disc Displacement/metabolism , Intervertebral Disc Displacement/pathology , Ligamentum Flavum/ultrastructure , Lumbar Vertebrae/pathology , Male , Microscopy, Electron , Middle Aged , Spinal Stenosis/pathology , Young Adult
5.
Braz J Med Biol Res ; 51(3): e7033, 2018 Jan 11.
Article in English | MEDLINE | ID: mdl-29340527

ABSTRACT

In the present study, we successfully demonstrated for the first time the existence of cardiac proteomic differences between non-selectively bred rats with distinct intrinsic exercise capacities. A proteomic approach based on two-dimensional gel electrophoresis coupled to mass spectrometry was used to study the left ventricle (LV) tissue proteome of rats with distinct intrinsic exercise capacity. Low running performance (LRP) and high running performance (HRP) rats were categorized by a treadmill exercise test, according to distance run to exhaustion. The running capacity of HRPs was 3.5-fold greater than LRPs. Protein profiling revealed 29 differences between HRP and LRP rats (15 proteins were identified). We detected alterations in components involved in metabolism, antioxidant and stress response, microfibrillar and cytoskeletal proteins. Contractile proteins were upregulated in the LVs of HRP rats (α-myosin heavy chain-6, myosin light chain-1 and creatine kinase), whereas the LVs of LRP rats exhibited upregulation in proteins associated with stress response (aldehyde dehydrogenase 2, α-crystallin B chain and HSPß-2). In addition, the cytoskeletal proteins desmin and α-actin were upregulated in LRPs. Taken together, our results suggest that the increased contractile protein levels in HRP rats partly accounted for their improved exercise capacity, and that proteins considered risk factors to the development of cardiovascular disease were expressed in higher amounts in LRP animals.


Subject(s)
Heart Function Tests/methods , Myocardium/metabolism , Physical Conditioning, Animal/physiology , Proteins/metabolism , Running/physiology , Animals , Contractile Proteins/metabolism , Cytoskeletal Proteins/metabolism , Desmin/metabolism , Electrophoresis, Gel, Two-Dimensional , Heart Ventricles/metabolism , Heat-Shock Proteins/metabolism , Male , Mass Spectrometry , Organ Size , Proteins/isolation & purification , Proteomics , Rats , Rats, Inbred Strains
6.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;51(3): e7033, 2018. tab, graf
Article in English | LILACS | ID: biblio-889046

ABSTRACT

In the present study, we successfully demonstrated for the first time the existence of cardiac proteomic differences between non-selectively bred rats with distinct intrinsic exercise capacities. A proteomic approach based on two-dimensional gel electrophoresis coupled to mass spectrometry was used to study the left ventricle (LV) tissue proteome of rats with distinct intrinsic exercise capacity. Low running performance (LRP) and high running performance (HRP) rats were categorized by a treadmill exercise test, according to distance run to exhaustion. The running capacity of HRPs was 3.5-fold greater than LRPs. Protein profiling revealed 29 differences between HRP and LRP rats (15 proteins were identified). We detected alterations in components involved in metabolism, antioxidant and stress response, microfibrillar and cytoskeletal proteins. Contractile proteins were upregulated in the LVs of HRP rats (α-myosin heavy chain-6, myosin light chain-1 and creatine kinase), whereas the LVs of LRP rats exhibited upregulation in proteins associated with stress response (aldehyde dehydrogenase 2, α-crystallin B chain and HSPβ-2). In addition, the cytoskeletal proteins desmin and α-actin were upregulated in LRPs. Taken together, our results suggest that the increased contractile protein levels in HRP rats partly accounted for their improved exercise capacity, and that proteins considered risk factors to the development of cardiovascular disease were expressed in higher amounts in LRP animals.


Subject(s)
Animals , Male , Rats , Physical Conditioning, Animal/physiology , Running/physiology , Proteins/metabolism , Heart Function Tests/methods , Myocardium/metabolism , Organ Size , Rats, Inbred Strains , Mass Spectrometry , Electrophoresis, Gel, Two-Dimensional , Proteins/isolation & purification , Contractile Proteins/metabolism , Cytoskeletal Proteins/metabolism , Proteomics , Desmin/metabolism , Heart Ventricles/metabolism , Heat-Shock Proteins/metabolism
7.
Cell Death Dis ; 8(11): e3166, 2017 11 09.
Article in English | MEDLINE | ID: mdl-29120410

ABSTRACT

Wallerian degeneration is an active program tightly associated with axonal degeneration, required for axonal regeneration and functional recovery after nerve damage. Here we provide a functional molecular foundation for our undertstanding of the complex non-cell autonomous role of glial cells in the regulation of axonal degeneration. To shed light on the complexity of the molecular machinery governing axonal degeneration we employ a multi-model, unbiased, in vivo approach combining morphological assesment and quantitative proteomics with in silico-based higher order functional clustering to genetically uncouple the intrinsic and extrinsic processes governing Wallerian degeneration. Highlighting a pivotal role for glial cells in the early stages fragmenting the axon by a cytokinesis-like process and a cell autonomous stage of axonal disintegration associated to mitochondrial dysfunction.


Subject(s)
Axons/metabolism , Neuroglia/metabolism , Animals , Cell Dedifferentiation/drug effects , Cells, Cultured , Contractile Proteins/antagonists & inhibitors , Contractile Proteins/genetics , Contractile Proteins/metabolism , Peptidyl-Prolyl Isomerase F , Cyclophilins/deficiency , Cyclophilins/genetics , Dactinomycin/pharmacology , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Sheath/physiology , Neuroglia/cytology , Proteomics , RNA Interference , Rats , Rats, Sprague-Dawley , Sciatic Nerve/drug effects , Sciatic Nerve/injuries , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology , rho-Associated Kinases/metabolism
8.
Sci Rep ; 6: 29003, 2016 06 30.
Article in English | MEDLINE | ID: mdl-27357373

ABSTRACT

Changes in vascular permeability occur during inflammation and the actin cytoskeleton plays a crucial role in regulating endothelial cell contacts and permeability. We demonstrated recently that the actin-binding protein cortactin regulates vascular permeability via Rap1. However, it is unknown if the actin cytoskeleton contributes to increased vascular permeability without cortactin. As we consistently observed more actin fibres in cortactin-depleted endothelial cells, we hypothesised that cortactin depletion results in increased stress fibre contractility and endothelial barrier destabilisation. Analysing the contractile machinery, we found increased ROCK1 protein levels in cortactin-depleted endothelium. Concomitantly, myosin light chain phosphorylation was increased while cofilin, mDia and ERM were unaffected. Secretion of the barrier-stabilising hormone adrenomedullin, which activates Rap1 and counteracts actomyosin contractility, was reduced in plasma from cortactin-deficient mice and in supernatants of cortactin-depleted endothelium. Importantly, adrenomedullin administration and ROCK1 inhibition reduced actomyosin contractility and rescued the effect on permeability provoked by cortactin deficiency in vitro and in vivo. Our data suggest a new role for cortactin in controlling actomyosin contractility with consequences for endothelial barrier integrity.


Subject(s)
Adrenomedullin/metabolism , Capillary Permeability/physiology , Cortactin/deficiency , Endothelial Cells/physiology , Actomyosin/physiology , Animals , Contractile Proteins/biosynthesis , Contractile Proteins/genetics , Cortactin/antagonists & inhibitors , Cortactin/genetics , Cortactin/physiology , Human Umbilical Vein Endothelial Cells , Humans , Lung/cytology , Male , Mice , RNA Interference , RNA, Small Interfering/genetics , Shelterin Complex , Telomere-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/physiology
9.
Thromb Res ; 138: 7-15, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26826502

ABSTRACT

INTRODUCTION: MAGP1 is a glycoprotein present in the elastic fibers and is a part of the microfibrils components. MAGP1 interacts with von Willebrand factor and the active form of TGF-ß and BMP. In mice lacking MAGP1, thrombus formation is delayed, increasing the occlusion time of carotid artery despite presenting normal blood coagulation in vitro. MAGP1-containing microfibrils may play a role in hemostasis and thrombosis. In this work, we evaluated the function of MAGP1 and its relation to TGF-ß in the arterial thrombosis process. METHODS AND RESULTS: We analyzed thrombus formation time in wild type and MAGP1-deficient mice comparing Rose Bengal and Ferric Chloride induced arterial lesion. The potential participation of TGF-ß in this process was accessed when we treated both wild type and MAGP1-deficient mice with losartan (an antihypertensive drug that decreases TGF-ß activity) or captopril (an angiotensin converting enzyme inhibitor that was used as a control antihypertensive drug). Besides, we evaluated thrombus embolization and the gelatinolytic activity in the arterial walls in vitro and ex vivo. Losartan and captopril were able to recover the thrombus formation time without changing blood pressure, activated partial thromboplastin time (aPTT), PT (prothrombin time), platelet aggregation and adhesion, but decreased gelatinase activity. CONCLUSIONS: Our results suggest that both treatments are effective in the prevention of the sub-endothelial ECM degradation, allowing the recovery of normal thrombus formation.


Subject(s)
Antihypertensive Agents/therapeutic use , Captopril/therapeutic use , Contractile Proteins/genetics , Extracellular Matrix Proteins/genetics , Losartan/therapeutic use , Thrombosis/drug therapy , Thrombosis/genetics , Animals , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Carotid Arteries/physiopathology , Contractile Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Gelatinases/metabolism , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Platelet Aggregation/drug effects , Platelet Function Tests , RNA Splicing Factors , Thrombosis/metabolism , Thrombosis/physiopathology , Transforming Growth Factor beta/metabolism
10.
Am J Med Genet A ; 152A(3): 726-31, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20186808

ABSTRACT

Melnick-Needles syndrome (MNS) (OMIM 309350) is a rare, X-linked dominant condition, caused by mutations in the filamin A gene (FLNA, on Xq28). In females, the syndrome presents with bone dysplasia and characteristic facial changes. Affected males may show two different phenotypes. One is similar to the female phenotype and is seen in children born to unaffected mothers and suggesting new mutations. Alternatively, males born to affected mothers have an embryonic or perinatally lethal disorder. It has been claimed that MNS constitutes part of a spectrum including frontometaphyseal dysplasia, otopalatodigital syndrome type 1 (OPD1) and otopalatodigital syndrome type 2 (OPD2). These conditions are produced by different mutations in the filamin A gene (FLNA). MNS is caused by three different mutations in FLNA exon 22, to date detected only in females. We describe the clinical manifestations and present the results of FLNA exon 22 mutations screening in two boys with the perinatally lethal form of MNS and their affected mothers. In order to obtain DNA amplification from paraffin-embedded tissues, we designed a new method based on hemi-nested PCR. One of the children (and his mother) had a previously undescribed mutation produced by a double SNP in the positions 3776 and 3777 of the gene and leading to an amino acid substitution (NP_001447:p.[Gly1176Asp]). The second child (and his mother) had an already known mutation (NP_001447.2:p[.Ser1199Leu]). This is the first report confirming the presence FLNA mutations in boys with the perinatally lethal phenotype of MNS. (


Subject(s)
Abnormalities, Multiple/genetics , Contractile Proteins/genetics , Genetic Diseases, X-Linked/genetics , Microfilament Proteins/genetics , Osteochondrodysplasias/genetics , Abnormalities, Multiple/pathology , Adult , Amino Acid Substitution , Base Sequence , DNA Mutational Analysis/methods , DNA Primers/genetics , Exons , Fatal Outcome , Female , Filamins , Genetic Diseases, X-Linked/pathology , Humans , Infant, Newborn , Male , Osteochondrodysplasias/pathology , Phenotype , Polymerase Chain Reaction/methods , Polymorphism, Single Nucleotide , Pregnancy , Syndrome
11.
Tissue Cell ; 42(1): 53-60, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19833367

ABSTRACT

In Megalobulimus abbreviatus, the ultrastructural features and the contractile proteins of columellar, pharyngeal and foot retractor muscles were studied. These muscles are formed from muscular fascicles distributed in different planes that are separated by connective tissue rich in collagen fibrils. These cells contain thick and thin filaments, the latter being attached to dense bodies, lysosomes, sarcoplasmic reticulum, caveolae, mitochondria and glycogen granules. Three types of muscle cells were distinguished: T1 cells displayed the largest amount of glycogen and an intermediate number of mitochondria, suggesting the highest anaerobic metabolism; T2 cells had the largest number of mitochondria and less glycogen, which suggests an aerobic metabolism; T3 cells showed intermediate glycogen volumes, suggesting an intermediate anaerobic metabolism. The myofilaments in the pedal muscle contained paramyosin measuring between 40 and 80nm in diameter. Western Blot muscle analysis showed a 46-kDa band that corresponds to actin and a 220-kDa band that corresponds to myosin filaments. The thick filament used in the electrophoresis showed a protein band of 100kDa in the muscles, which may correspond to paramyosin.


Subject(s)
Contractile Proteins/ultrastructure , Muscle, Striated/ultrastructure , Snails/ultrastructure , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Adaptation, Physiological/physiology , Animals , Collagen/metabolism , Collagen/ultrastructure , Connective Tissue/metabolism , Connective Tissue/ultrastructure , Contractile Proteins/analysis , Contractile Proteins/metabolism , Energy Metabolism/physiology , Feeding Behavior/physiology , Glycogen/metabolism , Glycolysis/physiology , Locomotion/physiology , Microscopy, Electron, Transmission , Mitochondria/metabolism , Mitochondria/ultrastructure , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscle, Striated/metabolism , Myosins/metabolism , Myosins/ultrastructure , Organelles/metabolism , Organelles/ultrastructure , Snails/metabolism , Species Specificity , Tropomyosin/metabolism , Tropomyosin/ultrastructure
12.
Blood ; 111(8): 4137-44, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18281502

ABSTRACT

Mice lacking the extracellular matrix protein microfibril-associated glycoprotein-1 (MAGP1) display delayed thrombotic occlusion of the carotid artery following injury as well as prolonged bleeding from a tail vein incision. Normal occlusion times were restored when recombinant MAGP1 was infused into deficient animals prior to vessel wounding. Blood coagulation was normal in these animals as assessed by activated partial thromboplastin time and prothrombin time. Platelet number was lower in MAGP1-deficient mice, but the platelets showed normal aggregation properties in response to various agonists. MAGP1 was not found in normal platelets or in the plasma of wild-type mice. In ligand blot assays, MAGP1 bound to fibronectin, fibrinogen, and von Willebrand factor, but von Willebrand factor was the only protein of the 3 that bound to MAGP1 in surface plasmon resonance studies. These findings show that MAGP1, a component of microfibrils and vascular elastic fibers, plays a role in hemostasis and thrombosis.


Subject(s)
Carotid Arteries/pathology , Contractile Proteins/deficiency , Extracellular Matrix Proteins/deficiency , Thrombosis/pathology , Animals , Bleeding Time , Blood Coagulation/drug effects , Blood Platelets/drug effects , Blood Pressure/drug effects , Carotid Arteries/drug effects , Carotid Arteries/physiopathology , Cattle , Contractile Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Fibrinogen/metabolism , Fibronectins/metabolism , Humans , Immunohistochemistry , Injections , Mice , Mice, Inbred C57BL , Platelet Function Tests , Protein Binding/drug effects , RNA Splicing Factors , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Surface Plasmon Resonance , von Willebrand Factor/metabolism
13.
Arch. cardiol. Méx ; Arch. cardiol. Méx;76(supl.4): S67-S75, oct.-dic. 2006.
Article in Spanish | LILACS | ID: lil-568131

ABSTRACT

Activation of cellular receptors by diverse stimuli induces dramatic changes in shape and function to respond to the new circumstances of the cell. This modified behavior depends on the reorganization of the peripheral actin meshwork. An outstanding example of these processes can be found in platelets, from which much of the information available on cytoskeletal function has been obtained. Among the many actin-crosslinking proteins like spectrin, fimbrin or alpha actinin, filamin a (FLNa) emerges as the one with the highest potential in initiating the polimerization of actin filaments (F-actin) during the formation of tridimensional actin gels. FLNa also links actin filaments to the cytosolic domain of many membrane glycoproteins in platelets through its C-terminal region. In addition to participating in cell shape changes, FLNa is a scaffoldding protein that recruits numerous proteins involved in a completely different set of functions, including signal transduction, gene transcription regulation, and receptor translocation; however, the physiological role of FLNa in these processes has remained elusive. The purpose of the present communication is to briefly describe the characteristics of the macromolecules able to interact with FLNa and to discuss a possible role of FLNa during the transduction of signals from those molecular elements in platelets.


Subject(s)
Animals , Humans , Blood Platelets/physiology , Contractile Proteins/physiology , Cytoskeletal Proteins/physiology , Microfilament Proteins/physiology , Platelet Activation , Platelet Membrane Glycoproteins/physiology , Receptors, Cell Surface/physiology , Signal Transduction , Actins/physiology , Contractile Proteins , Contractile Proteins , Cytoskeletal Proteins , Cytoskeletal Proteins , Drosophila , Integrins/physiology , Microfilament Proteins , Microfilament Proteins , Phosphorylation , Platelet Activation/physiology , Platelet Membrane Glycoproteins , Receptors, Cell Surface , Signal Transduction/physiology
14.
Epilepsia ; 47(1): 211-4, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16417552

ABSTRACT

PURPOSE: Familial periventricular heterotopia (PH) represents a disorder of neuronal migration resulting in multiple gray-matter nodules along the lateral ventricular walls. Prior studies have shown that mutations in the filamin A (FLNA) gene can cause PH through an X-linked dominant pattern. Heterozygotic female patients usually remain asymptomatic until the second or third decade of life, when they may have predominantly focal seizures, whereas hemizygotic male fetuses typically die in utero. Recent studies have also reported mutations in FLNA in male patients with PH who are cognitively normal. We describe PH in three male siblings with PH due to FLNA, severe developmental regression, and West syndrome. METHODS: The study includes the three affected brothers and their parents. Video-EEG recordings and magnetic resonance image (MRI) scanning were performed on all individuals. Mutations for FLNA were detected by using polymerase chain reaction (PCR) on genomic DNA followed by single-stranded conformational polymorphism (SSCP) analysis or sequencing. RESULTS: Two of the siblings are monozygotic twins, and all had West syndrome with hypsarrhythmia on EEG. MRI of the brain revealed periventricular nodules of cerebral gray-matter intensity, typical for PH. Mutational analyses demonstrated a cytosine-to-thymidine missense mutation (c. C1286T), resulting in a threonine-to-methionine amino acid substitution in exon 9 of the FLNA gene. CONCLUSIONS: The association between PH and West syndrome, to our knowledge, has not been previously reported. Males with PH have been known to harbor FLNA mutations, although uniformly, they either show early lethality or survive and have a normal intellect. The current studies show that FLNA mutations can cause periventricular heterotopia, developmental regression, and West syndrome in male patients, suggesting that this type of FLNA mutation may contribute to severe neurologic deficits.


Subject(s)
Brain Diseases/genetics , Choristoma/genetics , Contractile Proteins/genetics , Developmental Disabilities/genetics , Microfilament Proteins/genetics , Mutation/genetics , Spasms, Infantile/genetics , Brain Diseases/epidemiology , Brain Diseases/pathology , Cerebral Ventricles/pathology , Choristoma/epidemiology , Choristoma/pathology , DNA Mutational Analysis , Developmental Disabilities/epidemiology , Developmental Disabilities/pathology , Electroencephalography/statistics & numerical data , Female , Filamins , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/pathology , Genotype , Humans , Infant , Magnetic Resonance Imaging , Male , Mutation, Missense/genetics , Pedigree , Phenotype , Sex Factors , Spasms, Infantile/epidemiology , Videotape Recording
15.
Arch Biochem Biophys ; 446(2): 140-50, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16442073

ABSTRACT

Filamin is a phosphoprotein that organizes actin filaments into networks. We report that a purified C-terminal recombinant region of filamin is a suitable substrate for calcineurin in vitro. Furthermore, 1 microM cyclosporin A (CsA), a specific calcineurin inhibitor, reduced the dephosphorylation of the recombinant fragment in 293FT cells. Mutagenesis analysis showed that a dephosphorylation step occurred in Ser 2152, which was previously shown to provide resistance to calpain cleavage when endogenous PKA is activated. In contrast, phosphorylation of Ser 2152 was recently reported to be necessary for membrane dynamic changes. In this regard, we found that CsA protects filamin in platelets from calpain degradation. Results could be combined with available information in a single model, assuming that some of the peptide fragments released by calcineurin-regulated calpain action could mediate actions in downstream pathways, which may help to resolve the controversies reported on the role of filamin phosphorylation in actin dynamics.


Subject(s)
Blood Platelets/metabolism , Calcineurin/metabolism , Contractile Proteins/metabolism , Microfilament Proteins/metabolism , Actins/metabolism , Animals , Calcineurin Inhibitors , Calpain/metabolism , Cell Line , Cell Membrane/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclosporine/pharmacology , Filamins , Humans , Mice , Mutagenesis , Phosphorylation , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Serine/metabolism , Signal Transduction
16.
Arch Cardiol Mex ; 76 Suppl 4: S67-75, 2006.
Article in Spanish | MEDLINE | ID: mdl-17469336

ABSTRACT

Activation of cellular receptors by diverse stimuli induces dramatic changes in shape and function to respond to the new circumstances of the cell. This modified behavior depends on the reorganization of the peripheral actin meshwork. An outstanding example of these processes can be found in platelets, from which much of the information available on cytoskeletal function has been obtained. Among the many actin-crosslinking proteins like spectrin, fimbrin or alpha actinin, filamin a (FLNa) emerges as the one with the highest potential in initiating the polimerization of actin filaments (F-actin) during the formation of tridimensional actin gels. FLNa also links actin filaments to the cytosolic domain of many membrane glycoproteins in platelets through its C-terminal region. In addition to participating in cell shape changes, FLNa is a scaffoldding protein that recruits numerous proteins involved in a completely different set of functions, including signal transduction, gene transcription regulation, and receptor translocation; however, the physiological role of FLNa in these processes has remained elusive. The purpose of the present communication is to briefly describe the characteristics of the macromolecules able to interact with FLNa and to discuss a possible role of FLNa during the transduction of signals from those molecular elements in platelets.


Subject(s)
Blood Platelets/physiology , Contractile Proteins/physiology , Cytoskeletal Proteins/physiology , Microfilament Proteins/physiology , Platelet Activation , Platelet Membrane Glycoproteins/physiology , Receptors, Cell Surface/physiology , Signal Transduction , Actins/physiology , Animals , Contractile Proteins/chemistry , Contractile Proteins/metabolism , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Drosophila , Filamins , Humans , Integrins/physiology , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Phosphorylation , Platelet Activation/physiology , Platelet Membrane Glycoproteins/genetics , Receptors, Cell Surface/genetics , Signal Transduction/physiology
17.
Biochem Biophys Res Commun ; 338(3): 1527-36, 2005 Dec 23.
Article in English | MEDLINE | ID: mdl-16274663

ABSTRACT

EhABP-120 is the first filamin identified in the parasitic protozoan Entamoeba histolytica. Filamins are a family of cross-linking actin-binding proteins that promote a dynamic orthogonal web. They have been reported to interact directly with more than 30 cellular proteins and some phosphoinositides. The biochemical consequences of these interactions may have either positive or negative effects on the cross-linking function and also form a link between the cytoskeleton and plasma membrane. In this study, the EhABP-120 carboxy-terminal domain (END) was biochemically characterized. This domain was able to associate to 3-sulfate galactosyl ceramide, a new lipid target for a member of the filamin family. Also, the END domain was able to dimerize "in vitro." Molecular modeling analysis showed that the dimeric region is stabilized by a disulfide bond. Electrostatic and docking studies suggest that an electropositive concave pocket at the dimeric END domain interacts simultaneously with several sulfogalactose moieties of the sulfatide.


Subject(s)
Contractile Proteins/chemistry , Contractile Proteins/metabolism , Entamoeba histolytica/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Sulfoglycosphingolipids/metabolism , Amino Acid Sequence , Animals , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Membrane/metabolism , Contractile Proteins/genetics , Entamoeba histolytica/genetics , Filamins , Microfilament Proteins/genetics , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Alignment , Substrate Specificity , Transfection
18.
J Cardiovasc Pharmacol ; 44(6): 688-95, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15550789

ABSTRACT

Eugenol (EUG) acts as a calcium antagonist but effects on the contractile proteins could also occur. We investigated inotropic effects of EUG in rat left ventricular papillary muscles, measuring isometric force, time variables, and post rest potentiation and EUG actions on the effects of Ca2+ (0.62 to 2.5 mM) and isoproterenol (5 ng/ml), on myosin ATPase activity and on the calcium currents in single ventricular myocytes. EUG reduced tension and time variables without altering the sarcoplasmic reticulum activity increasing post-pause relative potentiation. Isoproterenol and Ca2+ counteract these negative inotropic effects. Tetanic tension diminished, but not the myosin ATPase activity suggesting an isolated sarcolemmal effect. EUG 0.1 mM decreased the Ca2+ current amplitude in the entire potential range tested and 0.5 mM almost completely blocked this inward current. Results suggested that EUG depresses force without affecting the contractile machinery and its action is the only dependent blockade of the calcium inward current.


Subject(s)
Eugenol/pharmacology , Heart/physiology , Myocardial Contraction/drug effects , Oils, Volatile/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Calcium Chloride/pharmacology , Cardiotonic Agents/pharmacology , Cell Separation , Contractile Proteins/drug effects , Electrophysiology , Heart/drug effects , In Vitro Techniques , Isometric Contraction/drug effects , Isoproterenol/pharmacology , Male , Myocardium/enzymology , Myosins/metabolism , Papillary Muscles/drug effects , Patch-Clamp Techniques , Rats
19.
Mol Cell Biochem ; 260(1-2): 49-53, 2004 May.
Article in English | MEDLINE | ID: mdl-15228085

ABSTRACT

A C-terminal region of human endothelial actin-binding protein-280 (ABP-280 or ABP, non-muscle filamin) was subcloned and efficiently expressed in a mammalian cells system as indicated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immunoblotting analysis. As predicted by the aminoacid sequence, the fragment, a 79 kD peptide (residues 1671-2361, plus 3.9 kD from an N-terminal fusion peptide included in the expression plasmid), contained the two potential cAMP-dependent protein kinase (PKA) phosphorylation sites (serine 2152 and threonine 2336) predicted to be present in this region of the molecule. Incubation of cells in the presence of cAMP-elevating agents enhanced 32P uptake into the fragment. Site-directed mutagenesis analysis indicated that serine 2152 is the unique substrate in the C-terminal region of ABP for endogenously activated PKA. The functional implications of phosphorylation of this residue, which belongs to a serine-proline motif, are discussed in terms of the role of filamin in cytoskeleton reorganization.


Subject(s)
Contractile Proteins/metabolism , Microfilament Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Binding Sites/genetics , Blotting, Western , Cell Line , Contractile Proteins/genetics , Cyclic AMP-Dependent Protein Kinases , Endothelium/chemistry , Filamins , Humans , Microfilament Proteins/genetics , Molecular Weight , Mutagenesis, Site-Directed , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation , Precipitin Tests , Recombinant Proteins/metabolism , Serine/genetics , Serine/metabolism
20.
FEMS Microbiol Lett ; 233(1): 91-6, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15043874

ABSTRACT

Giardia lamblia is a multiflagellar parasite and one of the earliest diverging eukaryotic cells. It possesses a complex cytoskeleton based on different groups of microtubular structures - a ventral adhesive disc, four pairs of flagella, a median body and funis. Centrin is an important member of the EF-hand family of calcium-binding proteins, and it is known to show calcium-sensitive contractile behaviour. In the present study, we performed an ultrastructural localization of centrin in G. lamblia using several monoclonal antibodies to centrin. Microtubular structures such as the basal bodies, all the flagella axonemes, the adhesive disc, funis, and the median bodies presented positive labelling to centrin. In addition, the dense rods also demonstrated positive labelling. These results show that centrin is located in key positions related to microtubules. The role of centrin in these dynamic regions is discussed.


Subject(s)
Calcium-Binding Proteins/analysis , Chromosomal Proteins, Non-Histone/analysis , Giardia lamblia/ultrastructure , Animals , Cell Nucleus/chemistry , Cell Nucleus/ultrastructure , Contractile Proteins/analysis , Cytoskeletal Proteins/analysis , Cytoskeleton/chemistry , Flagella/chemistry , Flagella/ultrastructure , Fluorescent Antibody Technique , Giardia lamblia/chemistry , Microscopy, Fluorescence , Microscopy, Immunoelectron , Microtubules/chemistry , Microtubules/ultrastructure , Organelles/chemistry , Organelles/ultrastructure , Protozoan Proteins/analysis
SELECTION OF CITATIONS
SEARCH DETAIL