Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
FASEB J ; 38(5): e23504, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38421271

ABSTRACT

The function of kidney podocytes is closely associated with actin cytoskeleton regulated by Rho small GTPases. Loss of actin-driven cell adhesions and processes is connected to podocyte dysfunction, proteinuria, and kidney diseases. FilGAP, a GTPase-activating protein for Rho small GTPase Rac1, is abundantly expressed in kidney podocytes, and its gene is linked to diseases in a family with focal segmental glomerulosclerosis. In this study, we have studied the role of FilGAP in podocytes in vitro. Depletion of FilGAP in cultured podocytes induced loss of actin stress fibers and increased Rac1 activity. Conversely, forced expression of FilGAP increased stress fiber formation whereas Rac1 activation significantly reduced its formation. FilGAP localizes at the focal adhesion (FA), an integrin-based protein complex closely associated with stress fibers, that mediates cell-extracellular matrix (ECM) adhesion, and FilGAP depletion decreased FA formation and impaired attachment to the ECM. Moreover, in unique podocyte cell cultures capable of inducing the formation of highly organized processes including major processes and foot process-like projections, FilGAP depletion or Rac1 activation decreased the formation of these processes. The reduction of FAs and process formations in FilGAP-depleted podocyte cells was rescued by inhibition of Rac1 or P21-activated kinase 1 (PAK1), a downstream effector of Rac1, and PAK1 activation inhibited their formations. Thus, FilGAP contributes to both cell-ECM adhesion and process formation of podocytes by suppressing Rac1/PAK1 signaling.


Subject(s)
Podocytes , Actins , Kidney , GTPase-Activating Proteins/genetics , Extracellular Matrix
2.
PLoS One ; 19(1): e0297443, 2024.
Article in English | MEDLINE | ID: mdl-38277391

ABSTRACT

There has been only limited success to differentiate adult stem cells into cardiomyocyte subtypes. In the present study, we have successfully induced beating atrial and ventricular cardiomyocytes from rat hair-follicle-associated pluripotent (HAP) stem cells, which are adult stem cells located in the bulge area. HAP stem cells differentiated into atrial cardiomyocytes in culture with the combination of isoproterenol, activin A, bone morphogenetic protein 4 (BMP4), basic fibroblast growth factor (bFGF), and cyclosporine A (CSA). HAP stem cells differentiated into ventricular cardiomyocytes in culture with the combination of activin A, BMP4, bFGF, inhibitor of Wnt production-4 (IWP4), and vascular endothelial growth factor (VEGF). Differentiated atrial cardiomyocytes were specifically stained for anti-myosin light chain 2a (MLC2a) antibody. Ventricular cardiomyocytes were specially stained for anti-myosin light chain 2v (MLC2v) antibody. Quantitative Polymerase Chain Reaction (qPCR) showed significant expression of MLC2a in atrial cardiomyocytes and MLC2v in ventricular cardiomyocytes. Both differentiated atrial and ventricular cardiomyocytes showed characteristic waveforms in Ca2+ imaging. Differentiated atrial and ventricular cardiomyocytes formed long myocardial fibers and beat as a functional syncytium, having a structure similar to adult cardiomyocytes. The present results demonstrated that it is possible to induce cardiomyocyte subtypes, atrial and ventricular cardiomyocytes, from HAP stem cells.


Subject(s)
Myocytes, Cardiac , Pluripotent Stem Cells , Rats , Animals , Myocytes, Cardiac/metabolism , Vascular Endothelial Growth Factor A/metabolism , Hair Follicle , Cell Differentiation , Dietary Supplements
3.
Front Physiol ; 14: 1287275, 2023.
Article in English | MEDLINE | ID: mdl-38124716

ABSTRACT

Introduction: Mobilization of intracellular insulin granules to the plasma membrane plays a crucial role in regulating insulin secretion. However, the regulatory mechanisms of this mobilization process have been poorly understood due to technical limitations. In this study, we propose a convenient approach for assessing intracellular insulin granule behavior based on single-molecule analysis of insulin granule membrane proteins labeled with Quantum dot fluorescent nanocrystals. Methods: This approach allows us to analyze intracellular insulin granule movement with subpixel accuracy at 33 fps. We tracked two insulin granule membrane proteins, phogrin and zinc transporter 8, fused to HaloTag in rat insulinoma INS-1 cells and, by evaluating the tracks with mean-square displacement, demonstrated the characteristic behavior of insulin granules. Results and discussion: Pharmacological perturbations of microtubules and F-actin affected insulin granule behavior on distinct modalities. Specifically, microtubule dynamics and F-actin positively and negatively regulate insulin granule behavior, respectively, presumably by modulating each different behavioral mode. Furthermore, we observed impaired insulin granule behavior and cytoskeletal architecture under chronic treatment of high concentrations of glucose and palmitate. Our approach provides detailed information regarding intracellular insulin granule mobilization and its pathophysiological implications. This study sheds new light on the regulatory mechanisms of intracellular insulin granule mobilization and has important implications for understanding the pathogenesis of diabetes.

4.
Cell Struct Funct ; 48(2): 161-174, 2023 Sep 23.
Article in English | MEDLINE | ID: mdl-37482421

ABSTRACT

Invadopodia are protrusive structures that mediate the extracellular matrix (ECM) degradation required for tumor invasion and metastasis. Rho small GTPases regulate invadopodia formation, but the molecular mechanisms of how Rho small GTPase activities are regulated at the invadopodia remain unclear. Here we have identified FilGAP, a GTPase-activating protein (GAP) for Rac1, as a negative regulator of invadopodia formation in tumor cells. Depletion of FilGAP in breast cancer cells increased ECM degradation and conversely, overexpression of FilGAP decreased it. FilGAP depletion promoted the formation of invadopodia with ECM degradation. In addition, FilGAP depletion and Rac1 overexpression increased the emergence of invadopodia induced by epidermal growth factor, whereas FilGAP overexpression suppressed it. Overexpression of GAP-deficient FilGAP mutant enhanced invadopodia emergence as well as FilGAP depletion. The pleckstrin-homology (PH) domain of FilGAP binds phosphatidylinositol 3,4-bisphosphate [PI(3,4)P2], which is distributed on membranes of the invadopodia. FilGAP localized to invadopodia in breast cancer cells on the ECM, but FilGAP mutant lacking PI(3,4)P2-binding showed low localization. Similarly, the decrease of PI(3,4)P2 production reduced the FilGAP localization. Our results suggest that FilGAP localizes to invadopodia through its PH domain binding to PI(3,4)P2 and down-regulates invadopodia formation by inactivating Rac1, inhibiting ECM degradation in invasive tumor cells.Key words: invadopodia, breast carcinoma, Rac1, FilGAP, PI(3,4)P2.


Subject(s)
Breast Neoplasms , Podosomes , Humans , Female , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Podosomes/metabolism , Podosomes/pathology , rho GTP-Binding Proteins/metabolism , Cell Line, Tumor , Extracellular Matrix/metabolism , Extracellular Matrix/pathology
5.
STAR Protoc ; 3(4): 101726, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36170109

ABSTRACT

Heterotypic endomembrane fusion between static GLUT4-containing vesicles and traveling transferrin receptor-containing endosomes triggers insulin-responsive translocation of the GLUT4 glucose transporter. Here, we provide a protocol for preparing BODIPY-based fluorescent sensor molecules allowing detection of heterotypic endomembrane fusion through dequenching via streptavidin-biotin binding and ratiometrically analyzing insulin-responsive events with live-cell imaging. Although this protocol is for evaluating specific fusion processes relating GLUT4 translocation, it is also applicable to assessing other processes so long as sensor molecules can properly label target molecules. For complete details on the use and execution of this protocol, please refer to Hatakeyama et al. (2022).


Subject(s)
Endosomes , Insulin , Endosomes/metabolism , Insulin/metabolism , Glucose Transporter Type 4/metabolism
6.
iScience ; 25(4): 104164, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35434546

ABSTRACT

Quantitative features of GLUT4 glucose transporter's behavior deep inside cells remain largely unknown. Our previous analyses with live-cell imaging of intracellular GLUT4 trafficking demonstrated two crucial early events responsible for triggering insulin-responsive translocation processes, namely, heterotypic fusion and liberation. To quantify the regulation, interrelationships, and dynamics of the initial events more accurately and comprehensively, we herein applied three analyses, each based on our distinct dual-color live-cell imaging approaches. With these approaches, heterotypic fusion was found to be the first trigger for insulin-responsive GLUT4 redistributions, preceding liberation, and to be critically regulated by Akt substrate of 160 kDa (AS160) and actin dynamics. In addition, demonstrating the subcellular regional dependence of GLUT4 dynamics revealed that liberated GLUT4 molecules are promptly incorporated into the trafficking itinerary of transferrin receptors. Our approaches highlight the physiological significance of endosomal "GLUT4 molecule trafficking" rather than "GLUT4 vesicle delivery" to the plasma membrane in response to insulin.

7.
Sci Rep ; 10(1): 9687, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32546738

ABSTRACT

Despite successful clinical application of non-equilibrium atmospheric pressure plasma (APP), the details of the molecular mechanisms underlying APP-inducible biological responses remain ill-defined. We previously reported that exposure of 3T3L1 cells to APP-irradiated buffer raised the cytoplasmic free Ca2+ ([Ca2+]i) concentration by eliciting Ca2+ influx in a manner sensitive to transient receptor potential (TRP) channel inhibitors. However, the precise identity of the APP-responsive channel molecule(s) remains unclear. In the present study, we aimed to clarify channel molecule(s) responsible for indirect APP-responsive [Ca2+]i rises. siRNA-mediated silencing experiments revealed that TRPA1 and TRPV1 serve as the major APP-responsive Ca2+ channels in 3T3L1 cells. Conversely, ectopic expression of either TRPA1 or TRPV1 in APP-unresponsive C2C12 cells actually triggered [Ca2+]i elevation in response to indirect APP exposure. Desensitization experiments using 3T3L1 cells revealed APP responsiveness to be markedly suppressed after pretreatment with allyl isothiocyanate or capsaicin, TRPA1 and TRPV1 agonists, respectively. APP exposure also desensitized the cells to these chemical agonists, indicating the existence of a bi-directional heterologous desensitization property of APP-responsive [Ca2+]i transients mediated through these TRP channels. Mutational analyses of key cysteine residues in TRPA1 (Cys421, Cys621, Cys641, and Cys665) and in TRPV1 (Cys258, Cys363, and Cys742) have suggested that multiple reactive oxygen and nitrogen species are intricately involved in activation of the channels via a broad range of modifications involving these cysteine residues. Taken together, these observations allow us to conclude that both TRPA1 and TRPV1 channels play a pivotal role in evoking indirect APP-dependent [Ca2+]i responses.


Subject(s)
Calcium/metabolism , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism , 3T3-L1 Cells , Animals , Atmospheric Pressure , Cell Line , Gene Knockdown Techniques , Humans , Mice , Myoblasts/metabolism , Real-Time Polymerase Chain Reaction
8.
FASEB J ; 33(9): 10240-10256, 2019 09.
Article in English | MEDLINE | ID: mdl-31211923

ABSTRACT

The neuropathological hallmarks of Parkinson's disease (PD) include the appearance of α-synuclein (α-SYN)-positive Lewy bodies (LBs) and the loss of catecholaminergic neurons. Thus, a potential mechanism promoting the uptake of extracellular α-SYN may exist in susceptible neurons. Of the various differentially expressed proteins, we are interested in flotillin (FLOT)-1 because this protein is highly expressed in the brainstem catecholaminergic neurons and is strikingly up-regulated in PD brains. In this study, we found that extracellular monomeric and fibrillar α-SYN can potentiate FLOT1-dopamine transporter (DAT) binding and pre-endocytic clustering of DAT on the cell surface, thereby facilitating DAT endocytosis and down-regulating its transporter activity. Moreover, we demonstrated that α-SYN itself exploited the DAT endocytic process to enter dopaminergic neuron-like cells, and both FLOT1 and DAT were found to be the components of LBs. Altogether, these findings revealed a novel role of extracellular α-SYN on cellular trafficking of DAT and may provide a rationale for the cell type-specific, functional, and pathologic alterations in PD.-Kobayashi, J., Hasegawa, T., Sugeno, N., Yoshida, S., Akiyama, T., Fujimori, K., Hatakeyama, H., Miki, Y., Tomiyama, A., Kawata, Y., Fukuda, M., Kawahata, I., Yamakuni, T., Ezura, M., Kikuchi, A., Baba, T., Takeda, A., Kanzaki, M., Wakabayashi, K., Okano, H., Aoki, M. Extracellular α-synuclein enters dopaminergic cells by modulating flotillin-1-assisted dopamine transporter endocytosis.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine/metabolism , Dopaminergic Neurons/pathology , Lewy Bodies/pathology , Membrane Proteins/metabolism , Parkinson Disease/pathology , alpha-Synuclein/metabolism , Aged , Aged, 80 and over , Brain/metabolism , Brain/pathology , Cell Membrane/metabolism , Dopamine Plasma Membrane Transport Proteins/genetics , Dopaminergic Neurons/metabolism , Endocytosis , Humans , Lewy Bodies/metabolism , Membrane Proteins/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Protein Transport , alpha-Synuclein/genetics
10.
J Biol Chem ; 294(4): 1161-1172, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30482843

ABSTRACT

AS160 and Tbc1d1 are key Rab GTPase-activating proteins (RabGAPs) that mediate release of static GLUT4 in response to insulin or exercise-mimetic stimuli, respectively, but their cooperative regulation and its underlying mechanisms remain unclear. By employing GLUT4 nanometry with cell-based reconstitution models, we herein analyzed the functional cooperative activities of the RabGAPs. When both RabGAPs are present, Tbc1d1 functionally dominates AS160, and stimuli-inducible GLUT4 release relies on Tbc1d1-evoking proximal stimuli, such as AICAR and intracellular Ca2+ Detailed functional assessments with varying expression ratios revealed that AS160 modulates sensitivity to external stimuli in Tbc1d1-mediated GLUT4 release. For example, Tbc1d1-governed GLUT4 release triggered by Ca2+ plus insulin occurred more efficiently than that in cells with little or no AS160. Series of mutational analyses revealed that these synergizing actions rely on the phosphotyrosine-binding 1 (PTB1) and calmodulin-binding domains of Tbc1d1 as well as key phosphorylation sites of both AS160 (Thr642) and Tbc1d1 (Ser237 and Thr596). Thus, the emerging cooperative governance relying on the multiple regulatory nodes of both Tbc1d1 and AS160, functioning together, plays a key role in properly deciphering biochemical signals into a physical GLUT4 release process in response to insulin, exercise, and the two in combination.


Subject(s)
GTPase-Activating Proteins/metabolism , Glucose Transporter Type 4/metabolism , Muscle, Skeletal/metabolism , 3T3-L1 Cells , Animals , GTPase-Activating Proteins/genetics , Glucose Transporter Type 4/genetics , Humans , Mice , Muscle, Skeletal/cytology , Phosphorylation , Protein Transport , Signal Transduction
11.
Methods Mol Biol ; 1814: 425-448, 2018.
Article in English | MEDLINE | ID: mdl-29956248

ABSTRACT

Here, we describe protocols for three-dimensional tracking of single quantum dot-conjugated molecules with nanometer accuracy in living cells using conventional fluorescence microscopy. The technique exploits out-of-focus images of single emitters combined with an automated pattern-recognition open-source software that fits the images with proper model functions to extract the emitter coordinates. We describe protocols for targeting quantum dots to both membrane components and cytosolic proteins.


Subject(s)
Imaging, Three-Dimensional , Microscopy, Fluorescence/methods , Quantum Dots/chemistry , Algorithms , Calibration , Cell Line, Tumor , Cell Survival , Cytosol/metabolism , Data Analysis , G(M1) Ganglioside/metabolism , Humans , Image Processing, Computer-Assisted , Membrane Proteins/metabolism , Staining and Labeling
12.
Cell Rep ; 23(8): 2354-2364, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29791847

ABSTRACT

Metabolic immunomodulation involving IL-1 has been investigated for unfavorable metabolic effects, including obesity, but a potentially favorable role for IL-1 remains unclear. Here, we find mechanistic interactions between working skeletal muscles and locally recruited neutrophils expressing IL-1ß, which supports muscle performance through priming exercise-dependent GLUT4 translocation. Thus, during exercise, both IL-1α/ß-deficient and neutrophil-depleted mice similarly exhibit increased fatigability associated with impaired muscle glucose homeostasis due to GLUT4 dysregulation. Deficiency of IL-1-producing neutrophils results in intrinsic abnormalities represented by aberrant Rac1 signaling and irregular GLUT4-storage vesicles, suggesting that these properties are maintained by local IL-1 produced by recruited neutrophils upon exercise, possibly on a daily basis. We propose that neutrophils are highly engaged in skeletal muscle performance via IL-1 regulation, which coordinates favorable inflammatory microenvironments supporting muscle glucose metabolism.


Subject(s)
Glucose Transporter Type 4/metabolism , Interleukin-1/metabolism , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , Neutrophils/metabolism , Animals , Carbohydrate Metabolism , Homeostasis , Interleukin-6/metabolism , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Physical Conditioning, Animal , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Walking , rac1 GTP-Binding Protein/metabolism
13.
Am J Physiol Regul Integr Comp Physiol ; 315(1): R90-R103, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29513560

ABSTRACT

Skeletal muscle regeneration after injury is a complex process involving interactions between inflammatory microenvironments and satellite cells. Interleukin (IL)-1 is a key mediator of inflammatory responses and exerts pleiotropic impacts on various cell types. Thus, we aimed to investigate the role of IL-1 during skeletal muscle regeneration. We herein show that IL-1α/ß-double knockout (IL-1KO) mice exhibit delayed muscle regeneration after cardiotoxin (CTX) injection, characterized by delayed infiltrations of immune cells accompanied by suppressed local production of proinflammatory factors including IL-6 and delayed increase of paired box 7 (PAX7)-positive satellite cells postinjury compared with those of wild-type (WT) mice. A series of in vitro experiments using satellite cells obtained from the IL-1KO mice unexpectedly revealed that IL-1KO myoblasts have impairments in terms of both proliferation and differentiation, both of which were reversed by exogenous IL-1ß administration in culture. Intriguingly, the delay in myogenesis was not attributable to the myogenic transcriptional program since MyoD and myogenin were highly upregulated in IL-1KO cells, instead appearing, at least in part, to be due to dysregulation of cellular fusion events, possibly resulting from aberrant actin regulatory systems. We conclude that IL-1 plays a positive role in muscle regeneration by coordinating the initial interactions among inflammatory microenvironments and satellite cells. Our findings also provide compelling evidence that IL-1 is intimately engaged in regulating the fundamental function of myocytes.


Subject(s)
Cell Proliferation , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Muscle Development , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Regeneration , Satellite Cells, Skeletal Muscle/metabolism , Toxins, Biological , Animals , Disease Models, Animal , Interleukin-1alpha/deficiency , Interleukin-1alpha/genetics , Interleukin-1beta/deficiency , Interleukin-1beta/genetics , Male , Mice, Inbred BALB C , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Diseases/chemically induced , Muscular Diseases/genetics , Muscular Diseases/pathology , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenin/genetics , Myogenin/metabolism , Satellite Cells, Skeletal Muscle/pathology , Signal Transduction , Stem Cell Niche , Time Factors
14.
Mol Pain ; 14: 1744806918757286, 2018.
Article in English | MEDLINE | ID: mdl-29353540

ABSTRACT

Muscle pain is a common condition that relates to various pathologies. Muscle overuse induces muscle pain, and neutrophils are key players in pain production. Neutrophils also play a central role in chronic pain by secreting interleukin (IL)-18. The aim of this study was to investigate the involvement of neutrophils and IL-18 in a mouse model of muscle pain. The right hind leg muscles of BALB/c mice were stimulated electrically to induce excessive muscle contraction. The left hind leg muscles were not stimulated. The pressure pain threshold, number of neutrophils, and IL-18 levels were investigated. Furthermore, the effects of the IL-18-binding protein and Brilliant Blue G on pain were investigated. In stimulated muscles, pressure pain thresholds decreased, and neutrophil and IL-18 levels increased compared with that in non-stimulated muscles. The administration of IL-18-binding protein and Brilliant Blue G attenuated hyperalgesia caused by excessive muscle contraction. These results suggest that increased IL-18 secretion from larger numbers of neutrophils elicits mechanical hyperalgesia.


Subject(s)
Interleukin-18/metabolism , Myalgia/pathology , Myalgia/physiopathology , Neutrophils/pathology , Nociception , Animals , Cell Count , Disease Models, Animal , Electric Stimulation , Hyperalgesia/pathology , Hyperalgesia/physiopathology , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-18/administration & dosage , Male , Mice, Inbred BALB C , Muscle Contraction/drug effects , Muscles/drug effects , Muscles/pathology , Muscles/physiopathology , Myalgia/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Nociception/drug effects , Pain Threshold , Pressure , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Rosaniline Dyes/pharmacology
15.
Tohoku J Exp Med ; 244(1): 15-24, 2018 01.
Article in English | MEDLINE | ID: mdl-29311489

ABSTRACT

Rotator cuff tears (RCTs) are a common shoulder problem in the elderly that can lead to both muscle atrophy and fatty infiltration due to less physical load. Satellite cells, quiescent cells under the basal lamina of skeletal muscle fibers, play a major role in muscle regeneration. However, the myogenic potency of human satellite cells in muscles with fatty infiltration is unclear due to the difficulty in isolating from small samples, and the mechanism of the progression of fatty infiltration has not been elucidated. The purpose of this study was to analyze the population of myogenic and adipogenic cells in disused supraspinatus (SSP) and intact subscapularis (SSC) muscles of the RCTs from the same patients using fluorescence-activated cell sorting. The microstructure of the muscle with fatty infiltration was observed as a whole mount condition under multi-photon microscopy. Myogenic differentiation potential and gene expression were evaluated in satellite cells. The results showed that the SSP muscle with greater fatty infiltration surrounded by collagen fibers compared with the SSC muscle under multi-photon microscopy. A positive correlation was observed between the ratio of muscle volume to fat volume and the ratio of myogenic precursor to adipogenic precursor. Although no difference was observed in the myogenic potential between the two groups in cell culture, satellite cells in the disused SSP muscle showed higher intrinsic myogenic gene expression than those in the intact SSC muscle. Our results indicate that satellite cells from the disused SSP retain sufficient potential of muscle growth despite the fatty infiltration.


Subject(s)
Adipose Tissue/pathology , Muscle Development , Rotator Cuff Injuries/pathology , Rotator Cuff/pathology , Satellite Cells, Skeletal Muscle/pathology , Adipogenesis , Aged , Cell Separation , Female , Flow Cytometry , Gene Expression Profiling , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Muscle Fibers, Skeletal/metabolism , Muscular Atrophy/pathology , Rotator Cuff Injuries/genetics , Satellite Cells, Skeletal Muscle/metabolism
16.
Sci Rep ; 8(1): 1499, 2018 01 24.
Article in English | MEDLINE | ID: mdl-29367680

ABSTRACT

Olfactory receptors (ORs) mediate olfactory chemo-sensation in OR neurons. Herein, we have demonstrated that the OR chemo-sensing machinery functions in pancreatic ß-cells and modulates insulin secretion. First, we found several OR isoforms, including OLFR15 and OLFR821, to be expressed in pancreatic islets and a ß-cell line, MIN6. Immunostaining revealed OLFR15 and OLFR821 to be uniformly expressed in pancreatic ß-cells. In addition, mRNAs of Olfr15 and Olfr821 were detected in single MIN6 cells. These results indicate that multiple ORs are simultaneously expressed in individual ß-cells. Octanoic acid, which is a medium-chain fatty acid contained in food and reportedly interacts with OLFR15, potentiated glucose-stimulated insulin secretion (GSIS), thereby improving glucose tolerance in vivo. GSIS potentiation by octanoic acid was confirmed in isolated pancreatic islets and MIN6 cells and was blocked by OLFR15 knockdown. While Gα olf expression was not detectable in ß-cells, experiments using inhibitors and siRNA revealed that the pathway dependent on phospholipase C-inositol triphosphate, rather than cAMP-protein kinase A, mediates GSIS potentiation via OLFR15. These findings suggest that the OR system in pancreatic ß-cells has a chemo-sensor function allowing recognition of environmental substances obtained from food, and potentiates insulin secretion in a cell-autonomous manner, thereby modulating systemic glucose metabolism.


Subject(s)
Glucose/metabolism , Insulin-Secreting Cells/chemistry , Insulin-Secreting Cells/drug effects , Insulin/metabolism , Receptors, Odorant/analysis , Animals , Cell Line , Gene Expression Profiling , Immunohistochemistry , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis , Receptors, Odorant/genetics
17.
J Physiol ; 595(16): 5603-5621, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28556933

ABSTRACT

KEY POINTS: Comprehensive imaging analyses of glucose transporter 4 (GLUT4) behaviour in mouse skeletal muscle was conducted. Quantum dot-based single molecule nanometry revealed that GLUT4 molecules in skeletal myofibres are governed by regulatory systems involving 'static retention' and 'stimulus-dependent liberation'. Vital imaging analyses and super-resolution microscopy-based morphometry demonstrated that insulin liberates the GLUT4 molecule from its static state by triggering acute heterotypic endomembrane fusion arising from the very small GLUT4-containing vesicles in skeletal myofibres. Prior exposure to exercise-mimetic stimuli potentiated this insulin-responsive endomembrane fusion event involving GLUT4-containing vesicles, suggesting that this endomembranous regulation process is a potential site related to the effects of exercise. ABSTRACT: Skeletal muscle is the major systemic glucose disposal site. Both insulin and exercise facilitate translocation of the glucose transporter glucose transporter 4 (GLUT4) via distinct signalling pathways and exercise also enhances insulin sensitivity. However, the trafficking mechanisms controlling GLUT4 mobilization in skeletal muscle remain poorly understood as a resuly of technical limitations. In the present study, which employs various imaging techniques on isolated skeletal myofibres, we show that one of the initial triggers of insulin-induced GLUT4 translocation is heterotypic endomembrane fusion arising from very small static GLUT4-containing vesicles with a subset of transferrin receptor-containing endosomes. Importantly, pretreatment with exercise-mimetic stimuli potentiated the susceptibility to insulin responsiveness, as indicated by these acute endomembranous activities. We also found that AS160 exhibited stripe-like localization close to sarcomeric α-actinin and that insulin induced a reduction of the stripe-like localization accompanying changes in its detergent solubility. The results of the present study thus provide a conceptual framework indicating that GLUT4 protein trafficking via heterotypic fusion is a critical feature of GLUT4 translocation in skeletal muscles and also suggest that the efficacy of the endomembranous fusion process in response to insulin is involved in the benefits of exercise.


Subject(s)
Endosomes/metabolism , Glucose Transporter Type 4/metabolism , Insulin/metabolism , Muscle, Skeletal/metabolism , Animals , Female , Male , Mice, Transgenic , Protein Transport
18.
Mol Biol Cell ; 28(1): 173-181, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28035048

ABSTRACT

Quantum dots (QDs) are a powerful tool for quantitatively analyzing dynamic cellular processes by single-particle tracking. However, tracking of intracellular molecules with QDs is limited by their inability to penetrate the plasma membrane and bind to specific molecules of interest. Although several techniques for overcoming these problems have been proposed, they are either complicated or inconvenient. To address this issue, in this study, we developed a simple, convenient, and nontoxic method for labeling intracellular molecules in cells using HaloTag technology and electroporation. We labeled intracellular myosin motors with this approach and tracked their movement within cells. By simultaneously imaging myosin movement and F-actin architecture, we observed that F-actin serves not only as a rail but also as a barrier for myosin movement. We analyzed the effect of insulin on the movement of several myosin motors, which have been suggested to regulate intracellular trafficking of the insulin-responsive glucose transporter GLUT4, but found no significant enhancement in myosin motor motility as a result of insulin treatment. Our approach expands the repertoire of proteins for which intracellular dynamics can be analyzed at the single-molecule level.


Subject(s)
Single-Cell Analysis/methods , 3T3-L1 Cells , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Cell Membrane/metabolism , Cytoplasm/metabolism , Electroporation , Glucose Transporter Type 4/metabolism , Mice , Microscopy, Fluorescence , Myosins/metabolism , Protein Transport , Quantum Dots
19.
Article in English | MEDLINE | ID: mdl-24109963

ABSTRACT

Intracellular trafficking is a critical process for cell physiology. Previous extensive studies employing biochemical and molecular biological approaches have provided qualitative information about intracellular trafficking, but we have little quantitative information due to technical limitations of these assays. We therefore developed a novel method for quantifying intracellular trafficking based on single molecule imaging with Quantum dot (QD) fluorescent nanocrystals and quantitatively described the trafficking properties of some recycling proteins. We herein first describe how to label intracellular molecules with QD which has no cell permeability and how to quantify intracellular trafficking, and then we detail the development of a novel experimental system allowing multi-color simultaneous single molecule imaging for analyzing the relationships of intracellular trafficking activities among multiple molecules having distinct trafficking properties. Finally, we document how we confirmed the reliability of our system by simultaneously analyzing the intracellular movements of two recycling protein, GLUT4 glucose transporter and transferrin receptor. Since impairment of intracellular trafficking has critical etiological roles in various late-onset diseases such as type 2 diabetes, our novel imaging system may be a powerful tool for developing next-generation biomedical devices for diagnostics and medical treatment based on intracellular trafficking.


Subject(s)
Intracellular Space/metabolism , Molecular Imaging/methods , 3T3-L1 Cells , Animals , Color , Glucose Transporter Type 4/metabolism , Manometry , Mice , Protein Transport , Quantum Dots/metabolism , Receptors, Transferrin/metabolism , Staining and Labeling
20.
Mol Cell Biol ; 33(15): 3036-49, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23732912

ABSTRACT

The mechanisms underlying the cellular entry of the HIV-1 Tat protein transduction domain (TatP) and the molecular information necessary to improve the transduction efficiency of TatP remain unclear due to the technical limitations for direct visualization of TatP's behavior in cells. Using confocal microscopy, total internal reflection fluorescence microscopy, and four-dimensional microscopy, we developed a single-molecule tracking assay for TatP labeled with quantum dots (QDs) to examine the kinetics of TatP initially and immediately before, at the beginning of, and immediately after entry into living cells. We report that even when the number of multivalent TatP (mTatP)-QDs bound to a cell was low, each single mTatP-QD first locally induced the cell's lateral transport machinery to move the mTatP-QD toward the center of the cell body upon cross-linking of heparan sulfate proteoglycans. The centripetal and lateral movements were linked to the integrity and flow of actomyosin and microtubules. Individual mTatP underwent lipid raft-mediated temporal confinement, followed by complete immobilization, which ultimately led to endocytotic internalization. However, bivalent TatP did not sufficiently promote either cell surface movement or internalization. Together, these findings provide clues regarding the mechanisms of TatP cell entry and indicate that increasing the valence of TatP on nanoparticles allows them to behave as cargo delivery nanomachines.


Subject(s)
Endocytosis , HIV Infections/virology , HIV-1/physiology , Host-Pathogen Interactions , Virus Internalization , tat Gene Products, Human Immunodeficiency Virus/analysis , tat Gene Products, Human Immunodeficiency Virus/metabolism , Actins/metabolism , HIV Infections/metabolism , HeLa Cells , Humans , Membrane Microdomains/metabolism , Membrane Microdomains/virology , Microscopy, Confocal/methods , Microtubules/metabolism , Microtubules/virology , Molecular Imaging/methods , Pseudopodia/metabolism , Pseudopodia/virology , Quantum Dots
SELECTION OF CITATIONS
SEARCH DETAIL
...