1
|
Ma X, Shi Y, Shang Z. Epiberberine Improves Hyperglycemia and Ameliorates Insulin Sensitivity in Type 2 Diabetic Mice. Nephrology (Carlton) 2025; 30:e14430. [PMID: 39888160 DOI: 10.1111/nep.14430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 12/26/2024] [Accepted: 12/31/2024] [Indexed: 02/01/2025]
Abstract
AIM Type 2 diabetes mellitus (T2DM) is a metabolic syndrome characterised by absolute or relative insufficiency of insulin secretion. The alkaloids from Rhizoma coptidis have potential hypoglycemic effects. Epiberberine (EPI), a protoberberine alkaloid extracted from Rhizome coptidis, has been found to regulate lipid metabolism. Our study aimed to investigate the antidiabetic effects of EPI on mice with T2DM, as well as its underlying mechanism. METHODS The T2DM model in mice was established using a combination of high-fat diet and streptozotocin. Animals were divided into the control, T2DM, EPI-low dose (50 mg/kg EPI), EPI-medium dose (100 mg/kg EPI), EPI-high dose (200 mg/kg EPI) and metformin (MTF) (200 mg/kg MTF) groups. Body weight, water/food intake, serum lipids, blood glucose tolerance, insulin sensitivity, histopathological alterations, insulin signalling pathway and inflammation-related pathways in each group were detected. RESULTS EPI significantly reduced blood glucose levels and water/food intake in T2DM mice. EPI reduced the levels of total cholesterol, total triglyceride, low-density lipoprotein cholesterol, aspartate aminotransferase and alanine aminotransferase, and elevated the levels of high-density lipoprotein cholesterol in serum. EPI effectively improved oral glucose tolerance, alleviated hepatic insulin resistance, decreased glycosylated haemoglobin levels and increased liver glycogen content. EPI ameliorated the histopathological alterations of skeletal muscle and liver in T2DM mice. EPI stimulated the insulin signalling pathway by increasing glucose transporter type 4 levels and activating insulin receptor substrate-1, phosphatidylinositol 3-kinase and protein kinase B in skeletal muscle and liver. EPI reduced the levels of proinflammatory cytokine in serum and inhibited the activation of mitogen-activated protein kinase signalling in skeletal muscle and liver of diabetic mice. CONCLUSION Overall, these data demonstrate that EPI alleviates the symptoms of T2DM, providing new insights into EPI as a therapeutic compound for the alleviation of T2DM.
Collapse
Affiliation(s)
- Xiaohong Ma
- Nephrology Department, Shenzhen Bao'an Authentic Tcm Therapy Hospital, Shenzhen, China
| | - Yufeng Shi
- Internal Medicine Department, Shenzhen Bao'an Authentic Tcm Therapy Hospital, Shenzhen, China
| | - Zhitao Shang
- Internal Medicine Department, Shenzhen Bao'an Authentic Tcm Therapy Hospital, Shenzhen, China
| |
Collapse
|
2
|
Woo JR, Bae SH, Wales TE, Engen JR, Lee J, Jang H, Park S. The serine phosphorylations in the IRS-1 PIR domain abrogate IRS-1 and IR interaction. Proc Natl Acad Sci U S A 2024; 121:e2401716121. [PMID: 38625937 PMCID: PMC11046688 DOI: 10.1073/pnas.2401716121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 03/16/2024] [Indexed: 04/18/2024] Open
Abstract
Serine phosphorylations on insulin receptor substrate 1 (IRS-1) by diverse kinases aoccur widely during obesity-, stress-, and inflammation-induced conditions in models of insulin resistance and type 2 diabetes. In this study, we define a region within the human IRS-1, which is directly C-terminal to the PTB domain encompassing numerous serine phosphorylation sites including Ser307 (mouse Ser302) and Ser312 (mouse 307) creating a phosphorylation insulin resistance (PIR) domain. We demonstrate that the IRS-1 PTB-PIR with its unphosphorylated serine residues interacts with the insulin receptor (IR) but loses the IR-binding when they are phosphorylated. Surface plasmon resonance studies further confirm that the PTB-PIR binds stronger to IR than just the PTB domain, and that phosphorylations at Ser307, Ser312, Ser315, and Ser323 within the PIR domain result in abrogating the binding. Insulin-responsive cells containing the mutant IRS-1 with all these four serines changed into glutamates to mimic phosphorylations show decreased levels of phosphorylations in IR, IRS-1, and AKT compared to the wild-type IRS-1. Hydrogen-deuterium exchange mass spectrometry experiments indicating the PIR domain interacting with the N-terminal lobe and the hinge regions of the IR kinase domain further suggest the possibility that the IRS-1 PIR domain protects the IR from the PTP1B-mediated dephosphorylation.
Collapse
Affiliation(s)
- Ju Rang Woo
- Division of Convergence Technology, New Drug Development Center, KBIOHealth, Cheongju28160, Republic of Korea
| | - Seung-Hyun Bae
- Division of Rare and Refractory Cancer, Research Institute, National Cancer Center,Goyang10408, Republic of Korea
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy,Goyang10408, Republic of Korea
| | - Thomas E. Wales
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA02115
| | - John R. Engen
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA02115
| | - Jongsoon Lee
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan31151, Republic of Korea
| | - Hyonchol Jang
- Division of Rare and Refractory Cancer, Research Institute, National Cancer Center,Goyang10408, Republic of Korea
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy,Goyang10408, Republic of Korea
| | - SangYoun Park
- School of Systems Biomedical Science, Soongsil University, Seoul06978, Republic of Korea
- Integrative Institute of Basic Sciences, Soongsil University, Seoul06978, Republic of Korea
| |
Collapse
|
3
|
Ageeva T, Rizvanov A, Mukhamedshina Y. NF-κB and JAK/STAT Signaling Pathways as Crucial Regulators of Neuroinflammation and Astrocyte Modulation in Spinal Cord Injury. Cells 2024; 13:581. [PMID: 38607020 PMCID: PMC11011519 DOI: 10.3390/cells13070581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/18/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Spinal cord injury (SCI) leads to significant functional impairments below the level of the injury, and astrocytes play a crucial role in the pathophysiology of SCI. Astrocytes undergo changes and form a glial scar after SCI, which has traditionally been viewed as a barrier to axonal regeneration and functional recovery. Astrocytes activate intracellular signaling pathways, including nuclear factor κB (NF-κB) and Janus kinase-signal transducers and activators of transcription (JAK/STAT), in response to external stimuli. NF-κB and STAT3 are transcription factors that play a pivotal role in initiating gene expression related to astrogliosis. The JAK/STAT signaling pathway is essential for managing secondary damage and facilitating recovery processes post-SCI: inflammation, glial scar formation, and astrocyte survival. NF-κB activation in astrocytes leads to the production of pro-inflammatory factors by astrocytes. NF-κB and STAT3 signaling pathways are interconnected: NF-κB activation in astrocytes leads to the release of interleukin-6 (IL-6), which interacts with the IL-6 receptor and initiates STAT3 activation. By modulating astrocyte responses, these pathways offer promising avenues for enhancing recovery outcomes, illustrating the crucial need for further investigation into their mechanisms and therapeutic applications in SCI treatment.
Collapse
Affiliation(s)
- Tatyana Ageeva
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.R.)
| | - Albert Rizvanov
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.R.)
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, 420111 Kazan, Russia
| | - Yana Mukhamedshina
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.R.)
- Department of Histology, Cytology and Embryology, Kazan State Medical University, 420012 Kazan, Russia
| |
Collapse
|
4
|
Martínez Báez A, Ayala G, Pedroza-Saavedra A, González-Sánchez HM, Chihu Amparan L. Phosphorylation Codes in IRS-1 and IRS-2 Are Associated with the Activation/Inhibition of Insulin Canonical Signaling Pathways. Curr Issues Mol Biol 2024; 46:634-649. [PMID: 38248343 PMCID: PMC10814773 DOI: 10.3390/cimb46010041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/23/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
Insulin receptor substrates 1 and 2 (IRS-1 and IRS-2) are signaling adaptor proteins that participate in canonical pathways, where insulin cascade activation occurs, as well as in non-canonical pathways, in which phosphorylation of substrates is carried out by a diverse array of receptors including integrins, cytokines, steroid hormones, and others. IRS proteins are subject to a spectrum of post-translational modifications essential for their activation, encompassing phosphorylation events in distinct tyrosine, serine, and threonine residues. Tyrosine residue phosphorylation is intricately linked to the activation of the insulin receptor cascade and its interaction with SH2 domains within a spectrum of proteins, including PI3K. Conversely, serine residue phosphorylation assumes a different function, serving to attenuate the effects of insulin. In this review, we have identified over 50 serine residues within IRS-1 that have been reported to undergo phosphorylation orchestrated by a spectrum of kinases, thereby engendering the activation or inhibition of different signaling pathways. Furthermore, we delineate the phosphorylation of over 10 distinct tyrosine residues at IRS-1 or IRS-2 in response to insulin, a process essential for signal transduction and the subsequent activation of PI3K.
Collapse
Affiliation(s)
- Anabel Martínez Báez
- Infection Disease Research Center, National Institute of Public Health, Cuernavaca 62100, Mexico; (A.M.B.); (G.A.); (A.P.-S.)
| | - Guadalupe Ayala
- Infection Disease Research Center, National Institute of Public Health, Cuernavaca 62100, Mexico; (A.M.B.); (G.A.); (A.P.-S.)
| | - Adolfo Pedroza-Saavedra
- Infection Disease Research Center, National Institute of Public Health, Cuernavaca 62100, Mexico; (A.M.B.); (G.A.); (A.P.-S.)
| | - Hilda M. González-Sánchez
- CONAHCYT—Infection Disease Research Center, National Institute of Public Health, Cuernavaca 62100, Mexico;
| | - Lilia Chihu Amparan
- Infection Disease Research Center, National Institute of Public Health, Cuernavaca 62100, Mexico; (A.M.B.); (G.A.); (A.P.-S.)
| |
Collapse
|
5
|
Sato Y, Yoshimura K, Matsuda K, Haraguchi T, Marumo A, Yamagishi M, Sato S, Ito K, Yajima J. Membrane-bound myosin IC drives the chiral rotation of the gliding actin filament around its longitudinal axis. Sci Rep 2023; 13:19908. [PMID: 37963943 PMCID: PMC10646037 DOI: 10.1038/s41598-023-47125-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/09/2023] [Indexed: 11/16/2023] Open
Abstract
Myosin IC, a single-headed member of the myosin I family, specifically interacts with anionic phosphatidylinositol 4,5-bisphosphate (PI[4,5]P2) in the cell membrane via the pleckstrin homology domain located in the myosin IC tail. Myosin IC is widely expressed and physically links the cell membrane to the actin cytoskeleton; it plays various roles in membrane-associated physiological processes, including establishing cellular chirality, lipid transportation, and mechanosensing. In this study, we evaluated the motility of full-length myosin IC of Drosophila melanogaster via the three-dimensional tracking of quantum dots bound to actin filaments that glided over a membrane-bound myosin IC-coated surface. The results revealed that myosin IC drove a left-handed rotational motion in the gliding actin filament around its longitudinal axis, indicating that myosin IC generated a torque perpendicular to the gliding direction of the actin filament. The quantification of the rotational motion of actin filaments on fluid membranes containing different PI(4,5)P2 concentrations revealed that the rotational pitch was longer at lower PI(4,5)P2 concentrations. These results suggest that the torque generated by membrane-bound myosin IC molecules can be modulated based on the phospholipid composition of the cell membrane.
Collapse
Affiliation(s)
- Yusei Sato
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| | - Kohei Yoshimura
- Department of Biology, Graduate School of Science, Chiba University, 1-33, Inage, Chiba, Japan
| | - Kyohei Matsuda
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| | - Takeshi Haraguchi
- Department of Biology, Graduate School of Science, Chiba University, 1-33, Inage, Chiba, Japan
| | - Akisato Marumo
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| | - Masahiko Yamagishi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| | - Suguru Sato
- Department of Biology, Graduate School of Science, Chiba University, 1-33, Inage, Chiba, Japan
| | - Kohji Ito
- Department of Biology, Graduate School of Science, Chiba University, 1-33, Inage, Chiba, Japan.
| | - Junichiro Yajima
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan.
- Komaba Institute for Science, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo, 153-8902, Japan.
- Research Center for Complex Systems Biology, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo, 153-8902, Japan.
| |
Collapse
|
6
|
SMOC2 promotes aggressive behavior of fibroblast-like synoviocytes in rheumatoid arthritis through transcriptional and post-transcriptional regulating MYO1C. Cell Death Dis 2022; 13:1035. [PMID: 36513634 PMCID: PMC9747908 DOI: 10.1038/s41419-022-05479-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022]
Abstract
Fibroblast-like synoviocytes (FLSs), play a key role in perpetuating synovial inflammation and bone erosion in rheumatoid arthritis (RA), however, the underlying mechanism(s) of RA FLSs activation and aggression remain unclear. Identifying endogenous proteins that selectively target FLSs is urgently needed. Here, we systematically identified that secreted modular calcium-binding protein 2 (SMOC2), was significantly increased in RA FLSs and synovial tissues. SMOC2 knockdown specifically regulated cytoskeleton remodeling and decreased the migration and invasion of RA FLSs. Mechanistically, cytoskeleton-related genes were significantly downregulated in RA FLSs with reduced SMOC2 expression, especially the motor protein myosin1c (MYO1C). SMOC2 controlled MYO1C expression by SRY-related high-mobility group box 4 (SOX4) and AlkB homolog 5 (ALKHB5) mediated-m6A modification through transcriptional and post-transcriptional regulation. Furthermore, intra-articular Ad-shRNA-SMOC2 treatment attenuated synovial inflammation as well as bone and cartilage erosion in rats with collagen-induced arthritis (CIA). Our findings suggest that increased SMOC2 expression in FLSs may contribute to synovial aggression and joint destruction in RA. SMOC2 may serve as a potential target against RA. SMOC2-mediated regulation of the synovial migration and invasion in RA FLSs. In RA FLSs, SMOC2 is significantly increased, leading to the increased level of MYO1C via SOX4-mediated transcriptional regulation and ALKBH5-mediated m6A modification, thereby causing cytoskeleton remodeling and promoting RA FLSs migration and invasion. The Figure was drawn by Figdraw.
Collapse
|
7
|
Matsui K, Emoto M, Fukuda N, Nomiyama R, Yamada K, Tanizawa Y. SNARE-binding protein synaptosomal-associated protein of 29 kDa (SNAP29) regulates the intracellular sequestration of glucose transporter 4 (GLUT4) vesicles in adipocytes. J Diabetes Investig 2022; 14:19-27. [PMID: 36181414 PMCID: PMC9807150 DOI: 10.1111/jdi.13912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 08/16/2022] [Accepted: 09/07/2022] [Indexed: 01/07/2023] Open
Abstract
AIMS/INTRODUCTION Insulin stimulates translocation of glucose transporter 4 (GLUT4) from the perinuclear location to the plasma membrane. In the unstimulated state, intracellular vesicles containing GLUT4 are sequestered into specialized storage vesicles that have come to be known as the insulin-responsive compartment (IRC). The IRC is a functional compartment in the perinuclear region that is a target of the insulin signaling cascade, although its precise nature is unclear. Here, we report a novel molecular mechanism facilitating formation of the IRC. MATERIALS AND METHODS We determined synaptosomal-associated protein of 29 kDa (SNAP29) by mass spectrometry to be an EH domain-containing protein 1 (EHD1)-binding protein. Then, its expression was confirmed by western blotting. Subcellular localization of SNAP29 was determined by immunofluorescent microscopy. Interactions between SNAP29 and syntaxins were determined by immunoprecipitation. We measured glucose uptake and GLUT4 translocation in 3T3-L1 adipocyte expressing SNAP29 or silencing SNAP29. RESULTS We found SNAP29 to be localized in the perinuclear region and to show partial co-localization with GLUT4 under basal conditions. We also found that SNAP29 binds to syntaxin6, a Qc-SNARE, in adipocytes. In SNAP29-expressing cells, vesicles containing GLUT4 were observed to aggregate around the perinuclear region. In contrast, when SNAP29 was silenced, perinuclear GLUT4 vesicles were dispersed throughout the cytosol. Insulin-stimulated glucose uptake was inhibited in both SNAP29-expressing and SNAP29-silenced cells. CONCLUSIONS These data suggest that SNAP29 sequesters and anchors GLUT4-containing vesicles in the perinuclear region, and might have a role in the biogenesis of the perinuclear IRC.
Collapse
Affiliation(s)
- Kumiko Matsui
- Department of Endocrinology, Metabolism, Hematological Sciences and TherapeuticsYamaguchi University Graduate School of MedicineUbeJapan
| | - Masahiro Emoto
- Department of Endocrinology, Metabolism, Hematological Sciences and TherapeuticsYamaguchi University Graduate School of MedicineUbeJapan,Emoto ClinicUbeJapan
| | - Naofumi Fukuda
- Department of Endocrinology, Metabolism, Hematological Sciences and TherapeuticsYamaguchi University Graduate School of MedicineUbeJapan
| | - Ryuta Nomiyama
- Department of Endocrinology, Metabolism, Hematological Sciences and TherapeuticsYamaguchi University Graduate School of MedicineUbeJapan
| | - Kyoko Yamada
- Department of Endocrinology, Metabolism, Hematological Sciences and TherapeuticsYamaguchi University Graduate School of MedicineUbeJapan
| | - Yukio Tanizawa
- Department of Endocrinology, Metabolism, Hematological Sciences and TherapeuticsYamaguchi University Graduate School of MedicineUbeJapan
| |
Collapse
|
8
|
The Actin Cytoskeleton Responds to Inflammatory Cues and Alters Macrophage Activation. Cells 2022; 11:cells11111806. [PMID: 35681501 PMCID: PMC9180445 DOI: 10.3390/cells11111806] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 02/01/2023] Open
Abstract
Much remains to be learned about the molecular mechanisms underlying a class of human disorders called actinopathies. These genetic disorders are characterized by loss-of-function mutations in actin-associated proteins that affect immune cells, leading to human immunopathology. However, much remains to be learned about how cytoskeletal dysregulation promotes immunological dysfunction. The current study reveals that the macrophage actin cytoskeleton responds to LPS/IFNγ stimulation in a biphasic manner that involves cellular contraction followed by cellular spreading. Myosin II inhibition by blebbistatin blocks the initial contraction phase and lowers iNOS protein levels and nitric oxide secretion. Conversely, conditional deletion of Arp2/3 complex in macrophages attenuates spreading and increases nitric oxide secretion. However, iNOS transcription is not altered by loss of myosin II or Arp2/3 function, suggesting post-transcriptional regulation of iNOS by the cytoskeleton. Consistent with this idea, proteasome inhibition reverses the effects of blebbistatin and rescues iNOS protein levels. Arp2/3-deficient macrophages demonstrate two additional phenotypes: defective MHCII surface localization, and depressed secretion of the T cell chemokine CCL22. These data suggest that interplay between myosin II and Arp2/3 influences macrophage activity, and potentially impacts adaptive-innate immune coordination. Disrupting this balance could have detrimental impacts, particularly in the context of Arp2/3-associated actinopathies.
Collapse
|
9
|
Park JE, Kang E, Han JS. HM-chromanone attenuates TNF-α-mediated inflammation and insulin resistance by controlling JNK activation and NF-κB pathway in 3T3-L1 adipocytes. Eur J Pharmacol 2022; 921:174884. [PMID: 35288193 DOI: 10.1016/j.ejphar.2022.174884] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/14/2022] [Accepted: 03/09/2022] [Indexed: 11/03/2022]
Abstract
Obesity is a major public health problem worldwide and causes inflammation and insulin resistance in adipose tissue. We investigated the ability of (E)-5-hydroxy-7-methoxy-3-(2'-hydroxybenzyl)-4-chromanone (HM-chromanone) isolated from Portulaca oleracea to attenuate the activation of inflammatory cytokines and signaling pathways associated with tumor necrosis factor (TNF)-α-mediated inflammation and insulin resistance in 3T3-L1 adipocytes. TNF-α triggers the release of inflammatory cytokines and activation of the mitogen-activated protein kinase and nuclear factor (NF)-κB signaling pathways. In this study, HM-chromanone inhibited the production of inflammatory cytokines and chemokines [TNF-α, interleukin (IL)-6, IL-1β, and monocyte chemoattractant protein 1] involved in inflammation and insulin resistance. Furthermore, TNF-α treatment increased c-Jun-NH2 terminal kinase (JNK) phosphorylation, whereas HM-chromanone significantly decreased JNK phosphorylation in a dose-dependent manner. TNF-α treatment increased the activation of inhibitor kappa B (IκB) kinase (IKK), IκBα, and NF-κBp65 compared with that of the control. However, HM-chromanone significantly blocked IKK, IκBα, and NF-κBp65 activation. Upon adipocyte stimulation with TNF-α, phosphorylated insulin receptor substrate (pIRS)-1 serine 307 levels increased and pIRS-1 tyrosine 612 levels decreased compared with those of the control. Upon treatment with HM-chromanone, serine 307 phosphorylation of IRS-1 was inhibited and tyrosine 612 phosphorylation of IRS-1 was increased. Thus, HM-chromanone improved TNF-α-mediated inflammation and insulin resistance by regulating JNK activation and the NF-κB pathway, thereby reducing inflammatory cytokine secretion and inhibiting serine phosphorylation of IRS-1 in the insulin signaling pathway. These results suggest the potential of HM-chromanone to improve inflammatory conditions and insulin resistance in adipocytes.
Collapse
Affiliation(s)
- Jea Eun Park
- Department of Food Science and Nutrition, Pusan National University, Busan, 46241, South Korea.
| | - Eunji Kang
- Department of Food Science and Nutrition, Pusan National University, Busan, 46241, South Korea.
| | - Ji Sook Han
- Department of Food Science and Nutrition, Pusan National University, Busan, 46241, South Korea.
| |
Collapse
|
10
|
Ikezaki M, Nishitsuji K, Matsumura K, Manabe S, Shibukawa Y, Wada Y, Ito Y, Ihara Y. C-Mannosylated tryptophan-containing WSPW peptide binds to actinin-4 and alters E-cadherin subcellular localization in lung epithelial-like A549 cells. Biochimie 2021; 192:136-146. [PMID: 34673139 DOI: 10.1016/j.biochi.2021.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 11/18/2022]
Abstract
The Trp-x-x-Trp (W-x-x-W) peptide motif, a consensus site for C-mannosylation, is the functional motif in cytokine type I receptors or thrombospondin type I repeat (TSR) superfamily proteins. W-x-x-W motifs are important for physiological and pathological functions of their parental proteins, but effects of C-mannosylation on protein functions remain to be elucidated. By using chemically synthesized WSPW peptides and C-mannosylated WSPW peptides (C-Man-WSPW), we herein investigated whether C-mannosylation of WSPW peptides confer additional biological functions to WSPW peptides. C-Man-WSPW peptide, but not non-mannosylated WSPW, reduced E-cadherin levels in A549 cells. Via peptide mass fingerprinting analysis, we identified actinin-4 as a C-Man-WSPW-binding protein in A549 cells. Actinin-4 partly co-localized with E-cadherin or β-catenin, despite no direct interaction between actinin-4 and E-cadherin. C-Man-WSPW reduced co-localization of E-cadherin and actinin-4; non-mannosylated WSPW had no effect on localization. In actinin-4-knockdown cells, E-cadherin was upregulated and demonstrated a punctate staining pattern in the cytoplasm, which suggests that actinin-4 regulated cell-surface E-cadherin localization. Thus, C-mannosylation of WSPW peptides is required for interaction with actinin-4 that subsequently alters expression and subcellular localization of E-cadherin and morphology of epithelial-like cells. Our results therefore suggest a regulatory role of C-mannosylation of the W-x-x-W motif in interactions between the motif and its binding partner and will thereby enhance understanding of protein C-mannosylation.
Collapse
Affiliation(s)
- Midori Ikezaki
- Department of Biochemistry, School of Medicine, Wakayama Medical University, Wakayama, 641-8509, Japan
| | - Kazuchika Nishitsuji
- Department of Biochemistry, School of Medicine, Wakayama Medical University, Wakayama, 641-8509, Japan.
| | - Ko Matsumura
- Department of Biochemistry, School of Medicine, Wakayama Medical University, Wakayama, 641-8509, Japan
| | - Shino Manabe
- Laboratory of Functional Molecule Chemistry, Pharmaceutical Department and Institute of Medicinal Chemistry, Hoshi University, Tokyo, 142-8501, Japan; Research Center for Pharmaceutical Development, Graduate School of Pharmaceutical Sciences & Faculty of Pharmaceutical Sciences, Tohoku University, Miyagi, 980-8578, Japan
| | - Yukinao Shibukawa
- Department of Molecular Medicine, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka, 594-1101, Japan
| | - Yoshinao Wada
- Department of Molecular Medicine, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka, 594-1101, Japan
| | - Yukishige Ito
- RIKEN Cluster for Pioneering Research, Saitama, 351-0198, Japan; Graduate School of Science, Osaka University, Osaka, 560-0043, Japan
| | - Yoshito Ihara
- Department of Biochemistry, School of Medicine, Wakayama Medical University, Wakayama, 641-8509, Japan.
| |
Collapse
|
11
|
Åslund A, Bokhari MH, Wetterdal E, Martin R, Knölker HJ, Bengtsson T. Myosin 1c: A novel regulator of glucose uptake in brown adipocytes. Mol Metab 2021; 53:101247. [PMID: 33965643 PMCID: PMC8182130 DOI: 10.1016/j.molmet.2021.101247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/26/2021] [Accepted: 05/03/2021] [Indexed: 12/02/2022] Open
Abstract
Objective The potential of brown adipose tissue (BAT) to influence energy homeostasis in animals and humans is encouraging as this tissue can increase fatty acid and glucose utilization to produce heat through uncoupling protein 1 (UCP1), but the actual mechanism of how the cell regulates glucose uptake is not fully understood. Myosin 1c (Myo1c) is an unconventional motor protein involved in several cellular processes, including insulin-mediated glucose uptake via GLUT4 vesicle fusion in white adipocytes, but its role in glucose uptake in BAT has not previously been investigated. Methods Using the specific inhibitor pentachloropseudilin (PClP), a neutralizing antibody assay, and siRNA, we examined the role of Myo1c in mechanisms leading to glucose uptake both in vitro in isolated mouse primary adipocytes and in vivo in mice. Results Our results show that inhibition of Myo1c removes insulin-stimulated glucose uptake in white adipocytes, while inducing glucose uptake in brown adipocytes, independent of GLUT4, by increasing the expression, translation, and translocation of GLUT1 to the plasma membrane. Inhibition of Myo1c leads to the activation of PKA and downstream substrates p38 and ATF-2, which are known to be involved in the expression of β-adrenergic genes. Conclusions Myo1c is a PKA repressor and regulates glucose uptake into BAT. Myo1c is a BAT-specific regulator of glucose uptake. Myo1c inhibition leads to increased expression, translation, and translocation of GLUT1. Myo1c inhibition results in increased activation of PKA and its downstream targets.
Collapse
Affiliation(s)
- Alice Åslund
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, SE-106 91, Stockholm, Sweden
| | - Muhammad Hamza Bokhari
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, SE-106 91, Stockholm, Sweden
| | - Erika Wetterdal
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, SE-106 91, Stockholm, Sweden
| | - René Martin
- Faculty of Chemistry, Technical University of Dresden, Bergstrasse 66, 01069, Dresden, Germany
| | - Hans-Joachim Knölker
- Faculty of Chemistry, Technical University of Dresden, Bergstrasse 66, 01069, Dresden, Germany
| | - Tore Bengtsson
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, SE-106 91, Stockholm, Sweden.
| |
Collapse
|
12
|
Chang TT, Lin LY, Chen JW. A Novel Resolution of Diabetes: C-C Chemokine Motif Ligand 4 Is a Common Target in Different Types of Diabetes by Protecting Pancreatic Islet Cell and Modulating Inflammation. Front Immunol 2021; 12:650626. [PMID: 33968046 PMCID: PMC8102776 DOI: 10.3389/fimmu.2021.650626] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/09/2021] [Indexed: 01/07/2023] Open
Abstract
Systemic inflammation is related to hyperglycemia in diabetes mellitus (DM). C-C chemokine motif ligand (CCL) 4 is upregulated in type 1 & type 2 DM patients. This study aimed to investigate if CCL4 could be a potential target to improve blood sugar control in different experimental DM models. Streptozotocin-induced diabetic mice, Leprdb /JNarl diabetic mice, and C57BL/6 mice fed a high fat diet were used as the type 1 DM, type 2 DM, and metabolic syndrome model individually. Mice were randomly assigned to receive an anti-CCL4 neutralizing monoclonal antibody. The pancreatic β-cells were treated with streptozotocin for in vitro experiments. In streptozotocin-induced diabetic mice, inhibition of CCL4 controlled blood sugar, increased serum insulin levels, increased islet cell proliferation and decreased pancreatic interleukin (IL)-6 expression. In the type 2 diabetes and metabolic syndrome models, CCL4 inhibition retarded the progression of hyperglycemia, reduced serum tumor necrosis factor (TNF)-α and IL-6 levels, and improved insulin resistance via reducing the phosphorylation of insulin receptor substrate-1 in skeletal muscle and liver tissues. CCL4 inhibition directly protected pancreatic β-cells from streptozotocin stimulation. Furthermore, CCL4-induced IL-6 and TNF-α expressions could be abolished by siRNA of CCR2/CCR5. In summary, direct inhibition of CCL4 protected pancreatic islet cells, improved insulin resistance and retarded the progression of hyperglycemia in different experimental models, suggesting the critical role of CCL4-related inflammation in the progression of DM. Future experiments may investigate if CCL4 could be a potential target for blood sugar control in clinical DM.
Collapse
MESH Headings
- Animals
- Blood Glucose/metabolism
- Cell Line
- Chemokine CCL4/immunology
- Chemokine CCL4/metabolism
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/metabolism
- Female
- Glucose Tolerance Test
- Humans
- Inflammation/immunology
- Inflammation/metabolism
- Insulin/blood
- Insulin/metabolism
- Insulin-Secreting Cells/immunology
- Insulin-Secreting Cells/metabolism
- Islets of Langerhans/cytology
- Islets of Langerhans/immunology
- Islets of Langerhans/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Pancreas/cytology
- Pancreas/metabolism
- Mice
Collapse
Affiliation(s)
- Ting-Ting Chang
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Department and Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Liang-Yu Lin
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jaw-Wen Chen
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Department and Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Healthcare and Services Center, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
13
|
Abstract
Myosins constitute a superfamily of actin-based molecular motor proteins that mediates a variety of cellular activities including muscle contraction, cell migration, intracellular transport, the formation of membrane projections, cell adhesion, and cell signaling. The 12 myosin classes that are expressed in humans share sequence similarities especially in the N-terminal motor domain; however, their enzymatic activities, regulation, ability to dimerize, binding partners, and cellular functions differ. It is becoming increasingly apparent that defects in myosins are associated with diseases including cardiomyopathies, colitis, glomerulosclerosis, neurological defects, cancer, blindness, and deafness. Here, we review the current state of knowledge regarding myosins and disease.
Collapse
|
14
|
Arif E, Solanki AK, Srivastava P, Rahman B, Tash BR, Holzman LB, Janech MG, Martin R, Knölker HJ, Fitzgibbon WR, Deng P, Budisavljevic MN, Syn WK, Wang C, Lipschutz JH, Kwon SH, Nihalani D. The motor protein Myo1c regulates transforming growth factor-β-signaling and fibrosis in podocytes. Kidney Int 2019; 96:139-158. [PMID: 31097328 DOI: 10.1016/j.kint.2019.02.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/07/2019] [Accepted: 02/14/2019] [Indexed: 01/19/2023]
Abstract
Transforming growth factor-β (TGF-β) is known to play a critical role in the pathogenesis of many progressive podocyte diseases. However, the molecular mechanisms regulating TGF-β signaling in podocytes remain unclear. Using a podocyte-specific myosin (Myo)1c knockout, we demonstrate whether Myo1c is critical for TGF-β-signaling in podocyte disease pathogenesis. Specifically, podocyte-specific Myo1c knockout mice were resistant to fibrotic injury induced by Adriamycin or nephrotoxic serum. Further, loss of Myo1c also protected from injury in the TGF-β-dependent unilateral ureteral obstruction mouse model of renal interstitial fibrosis. Mechanistic analyses showed that loss of Myo1c significantly blunted TGF-β signaling through downregulation of canonical and non-canonical TGF-β pathways. Interestingly, nuclear rather than the cytoplasmic Myo1c was found to play a central role in controlling TGF-β signaling through transcriptional regulation. Differential expression analysis of nuclear Myo1c-associated gene promoters showed that nuclear Myo1c targeted the TGF-β responsive gene growth differentiation factor (GDF)-15 and directly bound to the GDF-15 promoter. Importantly, GDF15 was found to be involved in podocyte pathogenesis, where GDF15 was upregulated in glomeruli of patients with focal segmental glomerulosclerosis. Thus, Myo1c-mediated regulation of TGF-β-responsive genes is central to the pathogenesis of podocyte injury. Hence, inhibiting this process may have clinical application in treating podocytopathies.
Collapse
Affiliation(s)
- Ehtesham Arif
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ashish K Solanki
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Pankaj Srivastava
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Bushra Rahman
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Brian R Tash
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lawrence B Holzman
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael G Janech
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA; College of Charleston, Charleston, South Carolina, USA
| | - René Martin
- Department of Chemistry, TU Dresden, Dresden, Germany
| | | | - Wayne R Fitzgibbon
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Peifeng Deng
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Milos N Budisavljevic
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Wing-Kin Syn
- Department of Gastroenterology & Hepatology, Medical University of South Carolina, Charleston, South Carolina, USA; Section of Gastroenterology, Ralph H Johnson VA Medical Center, Charleston, South Carolina, USA; Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, (UPV/EHU), Vizcaya, Spain
| | - Cindy Wang
- Department of Gastroenterology & Hepatology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Joshua H Lipschutz
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Sang-Ho Kwon
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, USA
| | - Deepak Nihalani
- Department of Medicine, Nephrology Division, Medical University of South Carolina, Charleston, South Carolina, USA.
| |
Collapse
|
15
|
Stafeev IS, Vorotnikov AV, Ratner EI, Menshikov MY, Parfyonova YV. Latent Inflammation and Insulin Resistance in Adipose Tissue. Int J Endocrinol 2017; 2017:5076732. [PMID: 28912810 PMCID: PMC5585607 DOI: 10.1155/2017/5076732] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/17/2017] [Indexed: 02/06/2023] Open
Abstract
Obesity is a growing problem in modern society and medicine. It closely associates with metabolic disorders such as type 2 diabetes mellitus (T2DM) and hepatic and cardiovascular diseases such as nonalcoholic fatty liver disease, atherosclerosis, myocarditis, and hypertension. Obesity is often associated with latent inflammation; however, the link between inflammation, obesity, T2DM, and cardiovascular diseases is still poorly understood. Insulin resistance is the earliest feature of metabolic disorders. It mostly develops as a result of dysregulated insulin signaling in insulin-sensitive cells, as compared to inactivating mutations in insulin receptor or signaling proteins that occur relatively rare. Here, we argue that inflammatory signaling provides a link between latent inflammation, obesity, insulin resistance, and metabolic disorders. We further hypothesize that insulin-activated PI3-kinase pathway and inflammatory signaling mediated by several IκB kinases may constitute negative feedback leading to insulin resistance at least in the fat tissue. Finally, we discuss perspectives for anti-inflammatory therapies in treating the metabolic diseases.
Collapse
Affiliation(s)
- I. S. Stafeev
- Russian Cardiology Research and Production Centre, Moscow 121552, Russia
- Faculty of Basic Medicine, M.V. Lomonosov Moscow State University, Moscow 119192, Russia
- *I. S. Stafeev:
| | - A. V. Vorotnikov
- Russian Cardiology Research and Production Centre, Moscow 121552, Russia
- M.V. Lomonosov Moscow State University Medical Center, Moscow 119192, Russia
| | - E. I. Ratner
- Russian Cardiology Research and Production Centre, Moscow 121552, Russia
- Endocrinology Research Centre, Moscow 117031, Russia
| | - M. Y. Menshikov
- Russian Cardiology Research and Production Centre, Moscow 121552, Russia
| | - Ye. V. Parfyonova
- Russian Cardiology Research and Production Centre, Moscow 121552, Russia
- Faculty of Basic Medicine, M.V. Lomonosov Moscow State University, Moscow 119192, Russia
| |
Collapse
|
16
|
Abstract
Myosin-I molecular motors are proposed to play various cellular roles related to membrane dynamics and trafficking. In this Cell Science at a Glance article and the accompanying poster, we review and illustrate the proposed cellular functions of metazoan myosin-I molecular motors by examining the structural, biochemical, mechanical and cell biological evidence for their proposed molecular roles. We highlight evidence for the roles of myosin-I isoforms in regulating membrane tension and actin architecture, powering plasma membrane and organelle deformation, participating in membrane trafficking, and functioning as a tension-sensitive dock or tether. Collectively, myosin-I motors have been implicated in increasingly complex cellular phenomena, yet how a single isoform accomplishes multiple types of molecular functions is still an active area of investigation. To fully understand the underlying physiology, it is now essential to piece together different approaches of biological investigation. This article will appeal to investigators who study immunology, metabolic diseases, endosomal trafficking, cell motility, cancer and kidney disease, and to those who are interested in how cellular membranes are coupled to the underlying actin cytoskeleton in a variety of different applications.
Collapse
Affiliation(s)
- Betsy B McIntosh
- Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA
| | - E Michael Ostap
- Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA
| |
Collapse
|
17
|
Catici DAM, Amos HE, Yang Y, van den Elsen JMH, Pudney CR. The red edge excitation shift phenomenon can be used to unmask protein structural ensembles: implications for NEMO-ubiquitin interactions. FEBS J 2016; 283:2272-84. [PMID: 27028374 DOI: 10.1111/febs.13724] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 03/29/2016] [Indexed: 02/02/2023]
Abstract
To understand complex molecular interactions, it is necessary to account for molecular flexibility and the available equilibrium of conformational states. Only a small number of experimental approaches can access such information. Potentially steady-state red edge excitation shift (REES) spectroscopy can act as a qualitative metric of changes to the protein free energy landscape (FEL) and the equilibrium of conformational states. First, we validate this hypothesis using a single Trp-containing protein, NF-κB essential modulator (NEMO). We provide detailed evidence from chemical denaturation studies, macromolecular crowding studies, and the first report of the pressure dependence of the REES effect. Combination of these data demonstrate that the REES effect can report on the 'ruggedness' of the FEL and we present a phenomenological model, based on realistic physical interpretations, for fitting steady-state REES data to allow quantification of this aspect of the REES effect. We test the conceptual framework we have developed by correlating findings from NEMO ligand-binding studies with the REES data in a range of NEMO-ligand binary complexes. Our findings shed light on the nature of the interaction between NEMO and poly-ubiquitin, suggesting that NEMO is differentially regulated by poly-ubiquitin chain length and that this regulation occurs via a modulation of the available equilibrium of conformational states, rather than gross structural change. This study therefore demonstrates the potential of REES as a powerful tool for tackling contemporary issues in structural biology and biophysics and elucidates novel information on the structure-function relationship of NEMO and key interaction partners.
Collapse
Affiliation(s)
- Dragana A M Catici
- Department of Biology and Biochemistry, Faculty of Science, University of Bath, UK
| | - Hope E Amos
- Department of Biology and Biochemistry, Faculty of Science, University of Bath, UK
| | - Yi Yang
- Department of Biology and Biochemistry, Faculty of Science, University of Bath, UK
| | | | - Christopher R Pudney
- Department of Biology and Biochemistry, Faculty of Science, University of Bath, UK
| |
Collapse
|
18
|
Structural Analysis of the Myo1c and Neph1 Complex Provides Insight into the Intracellular Movement of Neph1. Mol Cell Biol 2016; 36:1639-54. [PMID: 27044863 DOI: 10.1128/mcb.00020-16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/21/2016] [Indexed: 11/20/2022] Open
Abstract
The Myo1c motor functions as a cargo transporter supporting various cellular events, including vesicular trafficking, cell migration, and stereociliary movements of hair cells. Although its partial crystal structures were recently described, the structural details of its interaction with cargo proteins remain unknown. This study presents the first structural demonstration of a cargo protein, Neph1, attached to Myo1c, providing novel insights into the role of Myo1c in intracellular movements of this critical slit diaphragm protein. Using small angle X-ray scattering studies, models of predominant solution conformation of unliganded full-length Myo1c and Myo1c bound to Neph1 were constructed. The resulting structures show an extended S-shaped Myo1c with Neph1 attached to its C-terminal tail. Importantly, binding of Neph1 did not induce a significant shape change in Myo1c, indicating this as a spontaneous process or event. Analysis of interaction surfaces led to the identification of a critical residue in Neph1 involved in binding to Myo1c. Indeed, a point mutant from this site abolished interaction between Neph1 and Myo1c when tested in the in vitro and in live-cell binding assays. Live-cell imaging, including fluorescence recovery after photobleaching, provided further support for the role of Myo1c in intracellular vesicular movement of Neph1 and its turnover at the membrane.
Collapse
|
19
|
You DJ, Park CR, Furlong M, Koo O, Lee C, Ahn C, Seong JY, Hwang JI. Dimer of arfaptin 2 regulates NF-κB signaling by interacting with IKKβ/NEMO and inhibiting IKKβ kinase activity. Cell Signal 2015; 27:2173-81. [PMID: 26296658 DOI: 10.1016/j.cellsig.2015.08.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 08/15/2015] [Indexed: 12/30/2022]
Abstract
IκB kinases (IKKs) are a therapeutic target due to their crucial roles in various biological processes, including the immune response, the stress response, and tumor development. IKKs integrate various upstream signals that activate NF-κB by phosphorylating IκB and also regulate many proteins related to cell growth and metabolism. Although they function as a heteromeric complex comprised of kinase subunits and an adaptor, these kinases produce distinct cellular responses by phosphorylating different target molecules, suggesting that they may also be regulated in a subtype-specific manner. In this study, arfaptin 2 was identified as an IKKβ-specific binding partner. Interestingly, arfaptin 2 also interacted with NEMO. Domain mapping studies revealed that the C-terminal region, including the IKKβ HLH domain and the first coiled-coil NEMO region were respectively required for interactions with the arfaptin 2 N-terminal flexible region. Overexpression of arfaptin 2 inhibited tumor necrosis factor (TNF)-α-stimulated nuclear factor-κB (NF-κB) signaling, whereas downregulation of arfaptin 2 by small interfering RNA enhanced NF-κB activity. Dimerization of arfaptin 2 through the Bin-Amphiphysin-Rvs domain may be essential to inhibit activation of NF-κB through multimodal interactions with IKKβs or IKKβ/NEMO, as ectopic expression of the arfaptin 2 fragment responsible for IKK interactions did not change TNFα-stimulated NF-κB activation. These data indicate that arfaptin 2 is the first molecule to regulate NF-κB signaling by interacting with the functional IKK complex but not by direct inhibiting IKKβ phosphorylation.
Collapse
Affiliation(s)
- Dong-Joo You
- Graduate School of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Republic of Korea
| | - Cho Rong Park
- Graduate School of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Republic of Korea
| | - Michael Furlong
- Graduate School of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Republic of Korea
| | - Okjae Koo
- Samsung Biomedical Research Institute, 130 Samsung-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 433-803, Republic of Korea
| | - Cheolju Lee
- Life Sciences Division, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 136-791, Republic of Korea
| | - Curie Ahn
- Transplantation Research Institute, Cancer Research Institute, Seoul National University, Yongun-dong, Jongno-gu, Seoul 110-799, Republic of Korea
| | - Jae Young Seong
- Graduate School of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Republic of Korea
| | - Jong-Ik Hwang
- Graduate School of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Republic of Korea.
| |
Collapse
|
20
|
Lecat S, Matthes HWD, Pepperkok R, Simpson JC, Galzi JL. A Fluorescent Live Imaging Screening Assay Based on Translocation Criteria Identifies Novel Cytoplasmic Proteins Implicated in G Protein-coupled Receptor Signaling Pathways. Mol Cell Proteomics 2015; 14:1385-99. [PMID: 25759509 PMCID: PMC4424407 DOI: 10.1074/mcp.m114.046698] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 02/20/2015] [Indexed: 12/13/2022] Open
Abstract
Several cytoplasmic proteins that are involved in G protein-coupled receptor signaling cascades are known to translocate to the plasma membrane upon receptor activation, such as beta-arrestin2. Based on this example and in order to identify new cytoplasmic proteins implicated in the ON-and-OFF cycle of G protein-coupled receptor, a live-imaging screen of fluorescently labeled cytoplasmic proteins was performed using translocation criteria. The screening of 193 fluorescently tagged human proteins identified eight proteins that responded to activation of the tachykinin NK2 receptor by a change in their intracellular localization. Previously we have presented the functional characterization of one of these proteins, REDD1, that translocates to the plasma membrane. Here we report the results of the entire screening. The process of cell activation was recorded on videos at different time points and all the videos can be visualized on a dedicated website. The proteins BAIAP3 and BIN1, partially translocated to the plasma membrane upon activation of NK2 receptors. Proteins ARHGAP12 and PKM2 translocated toward membrane blebs. Three proteins that associate with the cytoskeleton were of particular interest : PLEKHH2 rearranged from individual dots located near the cell-substrate adhesion surface into lines of dots. The speriolin-like protein, SPATC1L, redistributed to cell-cell junctions. The Chloride intracellular Channel protein, CLIC2, translocated from actin-enriched plasma membrane bundles to cell-cell junctions upon activation of NK2 receptors. CLIC2, and one of its close paralogs, CLIC4, were further shown to respond with the same translocation pattern to muscarinic M3 and lysophosphatidic LPA receptors. This screen allowed us to identify potential actors in signaling pathways downstream of G protein-coupled receptors and could be scaled-up for high-content screening.
Collapse
Affiliation(s)
- Sandra Lecat
- From the ‡GPCRs, Pain and Inflammation Team, UMR7242, CNRS-University of Strasbourg, LabEx Medalis, 300 Bvd Sébastien Brant, 67412 Illkirch, France;
| | - Hans W D Matthes
- From the ‡GPCRs, Pain and Inflammation Team, UMR7242, CNRS-University of Strasbourg, LabEx Medalis, 300 Bvd Sébastien Brant, 67412 Illkirch, France
| | - Rainer Pepperkok
- §European Molecular Biology Laboratory, Advanced Light Microscopy Facility, Meyerhofstr 1, 69117 Heidelberg, Germany
| | - Jeremy C Simpson
- ¶¶School of Biology and Environmental Science and UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Jean-Luc Galzi
- From the ‡GPCRs, Pain and Inflammation Team, UMR7242, CNRS-University of Strasbourg, LabEx Medalis, 300 Bvd Sébastien Brant, 67412 Illkirch, France
| |
Collapse
|
21
|
Catici DAM, Horne JE, Cooper GE, Pudney CR. Polyubiquitin Drives the Molecular Interactions of the NF-κB Essential Modulator (NEMO) by Allosteric Regulation. J Biol Chem 2015; 290:14130-9. [PMID: 25866210 DOI: 10.1074/jbc.m115.640417] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Indexed: 11/06/2022] Open
Abstract
The NF-κB essential modulator (NEMO) is the master regulator of NF-κB signaling, controlling the immune and nervous systems. NEMO affects the activity of IκB kinase-β (IKKβ), which relieves the inhibition of the NF-κB transcriptional regulation machinery. Despite major effort, there is only a very sparse, phenomenological understanding of how NEMO regulates IKKβ and shows specificity in its large range of molecular interactions. We explore the key molecular interactions of NEMO using a molecular biophysics approach, incorporating rapid-mixing stopped-flow, high-pressure, and CD spectroscopies. Our study demonstrates that NEMO has a significant degree of native structural disorder and that molecular flexibility and ligand-induced conformational change are at the heart of the molecular interactions of NEMO. We found that long chain length, unanchored, linear polyubiquitin drives NEMO activity, enhancing the affinity of NEMO for IKKβ and the kinase substrate IκBα and promoting membrane association. We present evidence that unanchored polyubiquitin achieves this regulation by inducing NEMO conformational change by an allosteric mechanism. We combine our quantitative findings to give a detailed molecular mechanistic model for the activity of NEMO, providing insight into the molecular mechanism of NEMO activity with broad implications for the biological role of free polyubiquitin.
Collapse
Affiliation(s)
- Dragana A M Catici
- From the Department of Biology and Biochemistry, Faculty of Science, University of Bath, Bath BA2 7AY, United Kingdom
| | - James E Horne
- From the Department of Biology and Biochemistry, Faculty of Science, University of Bath, Bath BA2 7AY, United Kingdom
| | - Grace E Cooper
- From the Department of Biology and Biochemistry, Faculty of Science, University of Bath, Bath BA2 7AY, United Kingdom
| | - Christopher R Pudney
- From the Department of Biology and Biochemistry, Faculty of Science, University of Bath, Bath BA2 7AY, United Kingdom
| |
Collapse
|
22
|
Bhattacharyya S, Feferman L, Tobacman JK. Carrageenan Inhibits Insulin Signaling through GRB10-mediated Decrease in Tyr(P)-IRS1 and through Inflammation-induced Increase in Ser(P)307-IRS1. J Biol Chem 2015; 290:10764-74. [PMID: 25784556 DOI: 10.1074/jbc.m114.630053] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Indexed: 01/22/2023] Open
Abstract
Inflammation induced by exposure to the common food additive carrageenan leads to insulin resistance by increase in Ser(P)(307)-insulin receptor substrate 1 (IRS1) and subsequent decline in the insulin-stimulated increase in Ser(P)(473)-AKT. Inhibition of carrageenan-induced inflammation reversed the increase in Ser(P)(307)-IRS1 but did not completely reverse the carrageenan-induced decline in Ser(P)(473)-AKT. To identify the additional mechanism responsible for the decrease in Ser(P)(473)-AKT, studies were performed in human HepG2 cells and in C57BL/6J mice. Following carrageenan, expression of GRB10 (growth factor receptor-bound 10 protein), an adaptor protein that binds to the insulin receptor and inhibits insulin signaling, increased significantly. GRB10 silencing blocked the carrageenan-induced reduction of the insulin-stimulated increase in Tyr(P)-IRS1 and partially reversed the decline in Ser(P)(473)-AKT. The combination of GRB10 silencing with BCL10 silencing and the reactive oxygen species inhibitor Tempol completely reversed the decline in Ser(P)(473)-AKT. After carrageenan, GRB10 promoter activity was enhanced because of activation by GATA2. A direct correlation between Ser(P)(473)-AKT and Ser(P)(401)-GATA2 was evident, and inhibition of AKT phosphorylation by the PI3K inhibitor LY294002 blocked Ser(401)-GATA2 phosphorylation and the increase in GRB10 expression. Studies indicated that carrageenan inhibited insulin signaling by two mechanisms: through the inflammation-mediated increase in Ser(P)(307)-IRS1, a negative regulator of insulin signaling, and through a transcriptional mechanism leading to increase in GRB10 expression and GRB10-inhibition of Tyr(P)-IRS1, a positive regulator of insulin signaling. These mechanisms converge to inhibit the insulin-induced increase in Ser(P)(473)-AKT. They provide internal feedback, mediated by Ser(P)(473)-AKT, Ser(P)(401)-GATA2, and nuclear GATA2, which links the opposing effects of serine and tyrosine phosphorylations of IRS1 and can modulate insulin responsiveness.
Collapse
Affiliation(s)
- Sumit Bhattacharyya
- From the Department of Medicine, University of Illinois at Chicago and the Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| | - Leo Feferman
- From the Department of Medicine, University of Illinois at Chicago and the Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| | - Joanne K Tobacman
- From the Department of Medicine, University of Illinois at Chicago and the Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| |
Collapse
|
23
|
Carnagarin R, Dharmarajan AM, Dass CR. PEDF-induced alteration of metabolism leading to insulin resistance. Mol Cell Endocrinol 2015; 401:98-104. [PMID: 25462587 DOI: 10.1016/j.mce.2014.11.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 10/21/2014] [Accepted: 11/10/2014] [Indexed: 02/06/2023]
Abstract
Pigment epithelium-derived factor (PEDF) is an anti-angiogenic, immunomodulatory, and neurotrophic serine protease inhibitor protein. PEDF is evolving as a novel metabolic regulatory protein that plays a causal role in insulin resistance. Insulin resistance is the central pathogenesis of metabolic disorders such as obesity, type 2 diabetes mellitus, polycystic ovarian disease, and metabolic syndrome, and PEDF is associated with them. The current evidence suggests that PEDF administration to animals induces insulin resistance, whereas neutralisation improves insulin sensitivity. Inflammation, lipolytic free fatty acid mobilisation, and mitochondrial dysfunction are the proposed mechanism of PEDF-mediated insulin resistance. This review summarises the probable mechanisms adopted by PEDF to induce insulin resistance, and identifies PEDF as a potential therapeutic target in ameliorating insulin resistance.
Collapse
Affiliation(s)
- Revathy Carnagarin
- Curtin Biosciences Research Precinct, Bentley 6102, Australia; School of Pharmacy, Curtin University, Bentley 6102, Australia
| | - Arunasalam M Dharmarajan
- Curtin Biosciences Research Precinct, Bentley 6102, Australia; School of Biomedical Science, Curtin University, Bentley 6102, Australia
| | - Crispin R Dass
- Curtin Biosciences Research Precinct, Bentley 6102, Australia; School of Pharmacy, Curtin University, Bentley 6102, Australia.
| |
Collapse
|
24
|
Zhou J, Wang Q, Ding Y, Zou MH. Hypochlorous acid via peroxynitrite activates protein kinase Cθ and insulin resistance in adipocytes. J Mol Endocrinol 2015; 54:25-37. [PMID: 25381390 PMCID: PMC4261204 DOI: 10.1530/jme-14-0213] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We recently reported that genetic deletion of myeloperoxidase (MPO) alleviates obesity-related insulin resistance in mice in vivo. How MPO impairs insulin sensitivity in adipocytes is poorly characterized. As hypochlorous acid (HOCl) is a principal oxidant product generated by MPO, we evaluated the effects of HOCl on insulin signaling in adipocytes differentiated from 3T3-L1 cells. Exposure of 3T3-L1 adipocytes to exogenous HOCl (200 μmol/l) attenuated insulin-stimulated 2-deoxyglucose uptake, GLUT4 translocation, and insulin signals, including tyrosine phosphorylation of insulin receptor substrate 1 (IRS1) and phosphorylation of Akt. Furthermore, treatment with HOCl induced phosphorylation of IRS1 at serine 307, inhibitor κB kinase (IKK), c-Jun NH2-terminal kinase (JNK), and phosphorylation of PKCθ (PKCθ). In addition, genetic and pharmacological inhibition of IKK and JNK abolished serine phosphorylation of IRS1 and impairment of insulin signaling by HOCl. Furthermore, knockdown of PKCθ using siRNA transfection suppressed phosphorylation of IKK and JNK and consequently attenuated the HOCl-impaired insulin signaling pathway. Moreover, activation of PKCθ by peroxynitrite was accompanied by increased phosphorylation of IKK, JNK, and IRS1-serine 307. In contrast, ONOO(-) inhibitors abolished HOCl-induced phosphorylation of PKCθ, IKK, JNK, and IRS1-serine 307, as well as insulin resistance. Finally, high-fat diet (HFD)-induced insulin resistance was associated with enhanced phosphorylation of PKCθ, IKK, JNK, and IRS1 at serine 307 in white adipose tissues from WT mice, all of which were not found in Mpo knockout mice fed HFDs. We conclude that HOCl impairs insulin signaling pathway by increasing ONOO(-) mediated phosphorylation of PKCθ, resulting in phosphorylation of IKK/JNK and consequent serine phosphorylation of IRS1 in adipocytes.
Collapse
Affiliation(s)
- Jun Zhou
- Section of Molecular MedicineBSEB 306A, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Qilong Wang
- Section of Molecular MedicineBSEB 306A, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Ye Ding
- Section of Molecular MedicineBSEB 306A, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Ming-Hui Zou
- Section of Molecular MedicineBSEB 306A, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
25
|
Baldeón R. L, Weigelt K, de Wit H, Ozcan B, van Oudenaren A, Sempértegui F, Sijbrands E, Grosse L, Freire W, Drexhage HA, Leenen PJM. Decreased serum level of miR-146a as sign of chronic inflammation in type 2 diabetic patients. PLoS One 2014; 9:e115209. [PMID: 25500583 PMCID: PMC4264887 DOI: 10.1371/journal.pone.0115209] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 11/19/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND There is increasing evidence that chronic inflammation is an important determinant in insulin resistance and in the pathogenesis of type 2 diabetes (T2D). MicroRNAs constitute a newly discovered system of cell regulation and in particular two microRNAs (miR-146a and miR-155) have been described as regulators and biomarkers of inflammation. AIM To determine a putative association between the levels of miR-146a and miR-155 in serum of T2D patients, clinical parameters and serological indicators of inflammation. METHODS We performed quantitative Real Time PCR (qPCR) of microRNAs from serum (56 Ecuadorian T2D ambulatory patients and 40 non-diabetic controls). In addition, we evaluated T2D-related serum cytokines.chemokines and growth factors using a commercially available multi-analyte cytometric bead array system. We correlated outcomes to clinical parameters, including BMI, HbA1c and lipid state. RESULTS The Ecuadorian non-diabetic controls appeared as overweight (BMI>25: patients 85%, controls 82.5%) and as dyslipidemic (hypercholesterolemia: patients 60.7%, controls 67.5%) as the patients. The serum levels of miR-146a were significantly reduced in T2D patients as compared to these non-diabetic, but obese/dyslipidemic control group (mean patients 0.61, mean controls set at 1; p = 0.042), those of miR-155 were normal.The serum levels of both microRNAs correlated to each other (r = 0.478; p<0.001) and to leptin levels. The microRNAs did not correlate to BMI, glycemia and dyslipidemia.From the tested cytokines, chemokines and growth factors, we found IL-8 and HGF significantly raised in T2D patients versus non-diabetic controls (p = 0.011 and 0.023 respectively). CONCLUSIONS This study shows decreased serum anti-inflammatory miR-146a, increased pro-inflammatory IL-8 and increased HGF (a vascular/insular repair factor) as discriminating markers of failure of glucose control occurring on the background of obesity and dyslipidemia.
Collapse
Affiliation(s)
- Lucy Baldeón R.
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Immunology, Central University of Ecuador, Quito, Ecuador
| | - Karin Weigelt
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Harm de Wit
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Behiye Ozcan
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Adri van Oudenaren
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | | | - Eric Sijbrands
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Laura Grosse
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Wilma Freire
- Institute of Research in Health and Nutrition, University San Francisco de Quito, Quito, Ecuador
| | - Hemmo A. Drexhage
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Prometeo Program SENESCYT, Central University of Ecuador and Universidad de las Fuerzas Armadas, Quito, Ecuador
| | - Pieter J. M. Leenen
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
26
|
Human resistin inhibits myogenic differentiation and induces insulin resistance in myocytes. BIOMED RESEARCH INTERNATIONAL 2013; 2013:804632. [PMID: 23509781 PMCID: PMC3590612 DOI: 10.1155/2013/804632] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 12/06/2012] [Accepted: 12/21/2012] [Indexed: 01/07/2023]
Abstract
This study is aimed to investigate the effect of human resistin on myocyte differentiation and insulin resistance. The human resistin eukaryotic expression vector was stable transfected into C2C12 myocyte cells and was transiently transfected into COS7 cells. The effects of human resistin on cell proliferation, cell cycle, and myogenic differentiation of C2C12 cells were examined. Glucose uptake assays was performed on C2C12 myotubes by using [3H] 2-deoxy-D-glucose. The mRNA levels of insulin receptor (IR) and glucose transporter 4 (GLUT4) were evaluated by semiquantitative RT-PCR. Results showed by the C2C12 cells transfected with human resistin gene compared with that without transfecting gene are as follows: (1) cell proliferation was significantly promoted, (2) after inducing differentiation, the myotube's diameters and expression of desmin and myoglobin decreased, and (3) glucose uptake ratio was lowered and expression of IR and GLUT4 decreased. However, there was no significant difference in the glucose uptake ratio between C2C12 myotubes treated with a human resistin conditioned medium of COS7 cells and treated with control medium. These results suggest that maybe human resistin has not a direct role on insulin sensitivity of myocytes. However, maybe it impaired the insulin sensitivity of myocytes through suppressing myogenesis and stimulating proliferation of myoblasts.
Collapse
|
27
|
Bond LM, Brandstaetter H, Kendrick-Jones J, Buss F. Functional roles for myosin 1c in cellular signaling pathways. Cell Signal 2013; 25:229-35. [PMID: 23022959 PMCID: PMC3715701 DOI: 10.1016/j.cellsig.2012.09.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 09/24/2012] [Indexed: 02/01/2023]
Abstract
Cellular signaling pathways underlie the transfer of information throughout the cell and to adjoining cells and so govern most critical cellular functions. Increasing evidence points to the molecular motor myosin 1c as a prominent player in many signaling cascades, from the integrin-dependent signaling involved in cell migration to the signaling events underlying insulin resistance. Myosin 1c functions on these pathways both via an important role in regulating lipid raft recycling and also via direct involvement in signaling cascades. This review provides an overview of the functional involvement of myosin 1c in cellular signaling and discusses the possible potential for myosin 1c as a target for drug-based treatments for human diseases.
Collapse
Affiliation(s)
- Lisa M Bond
- Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, United Kingdom
| | | | | | | |
Collapse
|
28
|
Tiwari A, Jung JJ, Inamdar SM, Nihalani D, Choudhury A. The myosin motor Myo1c is required for VEGFR2 delivery to the cell surface and for angiogenic signaling. Am J Physiol Heart Circ Physiol 2012; 304:H687-96. [PMID: 23262137 DOI: 10.1152/ajpheart.00744.2012] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Vascular endothelial growth factor receptor-2 (VEGFR2) is a receptor tyrosine kinase that is expressed in endothelial cells and regulates angiogenic signal transduction under both physiological and pathological conditions. VEGFR2 turnover at the plasma membrane (PM) is regulated by its transport through endocytic and secretory transport pathways. Short-range cargo trafficking along actin filaments is commonly regulated by motor proteins of myosin superfamily. In the current study, performed in primary human endothelial cells, we demonstrate that unconventional myosin 1c (Myo1c; class I family member) regulates the localization of VEGFR2 at the PM. We further demonstrate that the recruitment of VEGFR2 to the PM and its colocalization with Myo1c and caveolin-1 occur in response to VEGF-A (VEGF) stimulation. In addition, VEGF-induced delivery of VEGFR2 to the cell surface requires Myo1c; surface VEGFR2 levels are reduced in the absence of Myo1c and, more importantly, are restored by the overexpression of wild-type but not mutant Myo1c. Subcellular density gradient fractionation revealed that partitioning of VEGFR2 into caveolin-1- and Myo1c-enriched membrane fractions is dependent on VEGF stimulation. Myo1c depletion resulted in increased VEGF-induced VEGFR2 transport to the lysosomes for degradation and was rescued by applying either brefeldin A, which blocks trafficking between the endoplasmic reticulum and the Golgi complex, or dynasore, an inhibitor of dynamin-mediated endocytosis. Myo1c depletion also reduced VEGF-induced VEGFR2 phosphorylation at Y1175 and phosphorylation-dependent activation of ERK1/2 and c-Src kinase, leading to reduced cell proliferation and cell migration. This is the first report demonstrating that Myo1c is an important mediator of VEGF-induced VEGFR2 delivery to the cell surface and plays a role in angiogenic signaling.
Collapse
Affiliation(s)
- Ajit Tiwari
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | |
Collapse
|
29
|
Greenberg MJ, Ostap EM. Regulation and control of myosin-I by the motor and light chain-binding domains. Trends Cell Biol 2012. [PMID: 23200340 DOI: 10.1016/j.tcb.2012.10.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Members of the myosin-I family of molecular motors are expressed in many eukaryotes, where they are involved in a multitude of critical processes. Humans express eight distinct members of the myosin-I family, making it the second largest family of myosins expressed in humans. Despite the high degree of sequence conservation in the motor and light chain-binding domains (LCBDs) of these myosins, recent studies have revealed surprising diversity of function and regulation arising from isoform-specific differences in these domains. Here we review the regulation of myosin-I function and localization by the motor and LCBDs.
Collapse
Affiliation(s)
- Michael J Greenberg
- The Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA
| | | |
Collapse
|
30
|
Christoffel DJ, Golden SA, Heshmati M, Graham A, Birnbaum S, Neve RL, Hodes GE, Russo SJ. Effects of inhibitor of κB kinase activity in the nucleus accumbens on emotional behavior. Neuropsychopharmacology 2012; 37:2615-23. [PMID: 22781845 PMCID: PMC3473326 DOI: 10.1038/npp.2012.121] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Inhibitor of κB kinase (IκK) has historically been studied in the context of immune response and inflammation, but recent evidence demonstrates that IκK activity is necessary and sufficient for regulation of neuronal function. Chronic social defeat stress of mice increases IκK activity in the nucleus accumbens (NAc) and this increase is strongly correlated to depression-like behaviors. Inhibition of IκK signaling results in a reversal of chronic social defeat stress-induced social avoidance behavior. Here, we more completely define the role of IκK in anxiety and depressive-like behaviors. Mice underwent stereotaxic microinjection of a herpes simplex virus expressing either green fluorescent protein, a constitutively active form of IκK (IκKca), or a dominant negative form of IκK into the NAc. Of all three experimental groups, only mice expressing IκKca show a behavioral phenotype. Expression of IκKca results in a decrease in the time spent in the non-periphery zones of an open field arena and increased time spent immobile during a forced swim test. No baseline differences in sucrose preference were observed, but following the acute swim stress we noted a marked reduction in sucrose preference. To determine whether IκK activity alters responses to other acute stressors, we examined behavior and spine morphology in mice undergoing an acute social defeat stress. We found that IκKca enhanced social avoidance behavior and promoted thin spine formation. These data show that IκK in NAc is a critical regulator of both depressive- and anxiety-like states and may do so by promoting the formation of immature excitatory synapses.
Collapse
Affiliation(s)
- Daniel J Christoffel
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, New York, NY, USA
| | - Sam A Golden
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, New York, NY, USA
| | - Mitra Heshmati
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, New York, NY, USA
| | - Ami Graham
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shari Birnbaum
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rachael L Neve
- McGovern Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Georgia E Hodes
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, New York, NY, USA
| | - Scott J Russo
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, New York, NY, USA,Fishberg Department of Neuroscience, Mount Sinai School of Medicine, One Gustave Levy Place, Icahn 10-26A, 10029-6574 New York, NY, USA, Tel: +212 659 5917, Fax: +212 659 8510, E-mail:
| |
Collapse
|
31
|
Fan Y, Eswarappa SM, Hitomi M, Fox PL. Myo1c facilitates G-actin transport to the leading edge of migrating endothelial cells. ACTA ACUST UNITED AC 2012; 198:47-55. [PMID: 22778278 PMCID: PMC3392929 DOI: 10.1083/jcb.201111088] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Addition of actin monomer (G-actin) to growing actin filaments (F-actin) at the leading edge generates force for cell locomotion. The polymerization reaction and its regulation have been studied in depth. However, the mechanism responsible for transport of G-actin substrate to the cell front is largely unknown; random diffusion, facilitated transport via myosin II contraction, local synthesis as a result of messenger ribonucleic acid localization, or F-actin turnover all might contribute. By tracking a photoactivatable, nonpolymerizable actin mutant, we show vectorial transport of G-actin in live migrating endothelial cells (ECs). Mass spectrometric analysis identified Myo1c, an unconventional F-actin-binding motor protein, as a major G-actin-interacting protein. The cargo-binding tail domain of Myo1c interacted with G-actin, and the motor domain was required for the transport. Local microinjection of Myo1c promoted G-actin accumulation and plasma membrane ruffling, and Myo1c knockdown confirmed its contribution to G-actin delivery to the leading edge and for cell motility. In addition, there is no obvious requirement for myosin II contractile-based transport of G-actin in ECs. Thus, Myo1c-facilitated G-actin transport might be a critical node for control of cell polarity and motility.
Collapse
Affiliation(s)
- Yi Fan
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | |
Collapse
|
32
|
Copps KD, White MF. Regulation of insulin sensitivity by serine/threonine phosphorylation of insulin receptor substrate proteins IRS1 and IRS2. Diabetologia 2012; 55:2565-2582. [PMID: 22869320 PMCID: PMC4011499 DOI: 10.1007/s00125-012-2644-8] [Citation(s) in RCA: 733] [Impact Index Per Article: 56.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 04/23/2012] [Indexed: 12/11/2022]
Abstract
The insulin receptor substrate proteins IRS1 and IRS2 are key targets of the insulin receptor tyrosine kinase and are required for hormonal control of metabolism. Tissues from insulin-resistant and diabetic humans exhibit defects in IRS-dependent signalling, implicating their dysregulation in the initiation and progression of metabolic disease. However, IRS1 and IRS2 are regulated through a complex mechanism involving phosphorylation of >50 serine/threonine residues (S/T) within their long, unstructured tail regions. In cultured cells, insulin-stimulated kinases (including atypical PKC, AKT, SIK2, mTOR, S6K1, ERK1/2 and ROCK1) mediate feedback (autologous) S/T phosphorylation of IRS, with both positive and negative effects on insulin sensitivity. Additionally, insulin-independent (heterologous) kinases can phosphorylate IRS1/2 under basal conditions (AMPK, GSK3) or in response to sympathetic activation and lipid/inflammatory mediators, which are present at elevated levels in metabolic disease (GRK2, novel and conventional PKCs, JNK, IKKβ, mPLK). An emerging view is that the positive/negative regulation of IRS by autologous pathways is subverted/co-opted in disease by increased basal and other temporally inappropriate S/T phosphorylation. Compensatory hyperinsulinaemia may contribute strongly to this dysregulation. Here, we examine the links between altered patterns of IRS S/T phosphorylation and the emergence of insulin resistance and diabetes.
Collapse
Affiliation(s)
- K D Copps
- Howard Hughes Medical Institute, Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, CLS 16020, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - M F White
- Howard Hughes Medical Institute, Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, CLS 16020, 300 Longwood Avenue, Boston, MA, 02115, USA.
| |
Collapse
|
33
|
König HG, Fenner BJ, Byrne JC, Schwamborn RF, Bernas T, Jefferies CA, Prehn JHM. Fibroblast growth factor homologous factor 1 interacts with NEMO to regulate NF-κB signaling in neurons. J Cell Sci 2012; 125:6058-70. [DOI: 10.1242/jcs.111880] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Neuronal survival and plasticity critically depend on constitutive activity of the transcription factor nuclear factor-κB (NF-κB). We here describe a role for a small intracellular fibroblast growth factor homologue, the fibroblast growth factor homologous factor 1 (FHF1/FGF12) in the regulation of NF-κB activity in mature neurons. FHF's have previously been described to control neuronal excitability, and mutations in FHF isoforms give rise to a form of progressive spinocerebellar ataxia. Using a protein-array approach, we identified FHF1b as a novel interactor of the canonical NF–κB modulator IKKγ/NEMO. Co-immunoprecipitation, pull-down and GAL4-reporter experiments, as well as proximity ligation assays confirmed the interaction of FHF1 and NEMO, and demonstrated that a major site of interaction occurred within the axon initial segment. Fhf1 gene silencing strongly activated neuronal NF-κB activity and increased neurite lengths, branching patterns and spine counts in mature cortical neurons. The effects of FHF1 on neuronal NF-κB activity and morphology required the presence of NEMO. Our results imply that FHF1 negatively regulates the constitutive NF-κB activity in neurons.
Collapse
|
34
|
Jiang S, Messina JL. Role of inhibitory κB kinase and c-Jun NH2-terminal kinase in the development of hepatic insulin resistance in critical illness diabetes. Am J Physiol Gastrointest Liver Physiol 2011; 301:G454-63. [PMID: 21680774 PMCID: PMC3174535 DOI: 10.1152/ajpgi.00148.2011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hyperglycemia and insulin resistance induced by acute injuries or critical illness are associated with increased mortality and morbidity, as well as later development of type 2 diabetes. The molecular mechanisms underlying the acute onset of insulin resistance following critical illness remain poorly understood. In the present studies, the roles of serine kinases, inhibitory κB kinase (IKK) and c-Jun NH(2)-terminal kinase (JNK), in the acute development of hepatic insulin resistance were investigated. In our animal model of critical illness diabetes, activation of hepatic IKK and JNK was observed as early as 15 min, concomitant with the rapid impairment of hepatic insulin signaling and increased serine phosphorylation of insulin receptor substrate 1. Inhibition of IKKα or IKKβ, or both, by adenovirus vector-mediated expression of dominant-negative IKKα or IKKβ in liver partially restored insulin signaling. Similarly, inhibition of JNK1 kinase by expression of dominant-negative JNK1 also resulted in improved hepatic insulin signaling, indicating that IKK and JNK1 kinases contribute to critical illness-induced insulin resistance in liver.
Collapse
Affiliation(s)
- Shaoning Jiang
- 1Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, and
| | - Joseph L. Messina
- 1Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, and ,2Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
35
|
Abstract
NF-κB transcription factors are critical regulators of immunity, stress responses, apoptosis and differentiation. A variety of stimuli coalesce on NF-κB activation, which can in turn mediate varied transcriptional programs. Consequently, NF-κB-dependent transcription is not only tightly controlled by positive and negative regulatory mechanisms but also closely coordinated with other signaling pathways. This intricate crosstalk is crucial to shaping the diverse biological functions of NF-κB into cell type- and context-specific responses.
Collapse
|
36
|
Yoshida H, Kitagishi Y, Okumura N, Murakami M, Nishimura Y, Matsuda S. How do you RUN on? FEBS Lett 2011; 585:1707-10. [PMID: 21570977 DOI: 10.1016/j.febslet.2011.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 05/03/2011] [Accepted: 05/03/2011] [Indexed: 10/18/2022]
Abstract
RUN domain is present in several proteins related to the functions of Rap and Rab family GTPases. Accumulating evidence supports the hypothesis that RUN domain-containing proteins act as a component of vesicle traffic and might be responsible for an interaction with a filamentous network linked to actin cytoskeleton or microtubules. That is to say, on one hand, RUN domains associate with Rab or Rap family proteins, on the other hand, they also might interact with motor proteins such as kinesin or myosin via intervention molecules. In this review, we summarize the background and current status of RUN domain research with an emphasis on the interaction between RUN domain and motor proteins with respect to the vesicle traffic on filamentous network.
Collapse
Affiliation(s)
- Hitomi Yoshida
- Department of Environmental Health Science, Nara Women's University, Kita-Uoya Nishimachi, Nara, Japan
| | | | | | | | | | | |
Collapse
|
37
|
Pyrpassopoulos S, Shuman H, Ostap EM. Single-molecule adhesion forces and attachment lifetimes of myosin-I phosphoinositide interactions. Biophys J 2011; 99:3916-22. [PMID: 21156133 DOI: 10.1016/j.bpj.2010.10.043] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 10/10/2010] [Accepted: 10/27/2010] [Indexed: 11/19/2022] Open
Abstract
Phosphoinositides regulate the activities and localization of many cytoskeletal proteins involved in crucial biological processes, including membrane-cytoskeleton adhesion. Yet little is known about the mechanics of protein-phosphoinositide interactions, or about the membrane-attachment mechanics of any peripheral membrane proteins. Myosin-Ic (myo1c) is a molecular motor that links membranes to the cytoskeleton via phosphoinositide binding, so it is particularly important to understand the mechanics of its membrane attachment. We used optical tweezers to measure the strength and attachment lifetime of single myo1c molecules as they bind beads coated with a bilayer of 2% phosphatidylinositol 4,5-bisphosphate and 98% phosphatidylcholine. Adhesion forces measured under ramp-load ranged between 5.5 and 16 pN at loading rates between 250 and 1800 pN/s. Dissociation rates increased linearly with constant force (0.3-2.5 pN), with rates exceeding 360 s(-1) at 2.5 pN. Attachment lifetimes calculated from adhesion force measurements were loading-rate-dependent, suggesting nonadiabatic behavior during pulling. The adhesion forces of myo1c with phosphoinositides are greater than the motors stall forces and are within twofold of the force required to extract a lipid molecule from the membrane. However, attachment durations are short-lived, suggesting that phosphoinositides alone do not provide the mechanical stability required to anchor myo1c to membranes during multiple ATPase cycles.
Collapse
Affiliation(s)
- Serapion Pyrpassopoulos
- Pennsylvania Muscle Institute and Department of Physiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
38
|
Motor protein Myo1c is a podocyte protein that facilitates the transport of slit diaphragm protein Neph1 to the podocyte membrane. Mol Cell Biol 2011; 31:2134-50. [PMID: 21402783 DOI: 10.1128/mcb.05051-11] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The podocyte proteins Neph1 and nephrin organize a signaling complex at the podocyte cell membrane that forms the structural framework for a functional glomerular filtration barrier. Mechanisms regulating the movement of these proteins to and from the membrane are currently unknown. This study identifies a novel interaction between Neph1 and the motor protein Myo1c, where Myo1c plays an active role in targeting Neph1 to the podocyte cell membrane. Using in vivo and in vitro experiments, we provide data supporting a direct interaction between Neph1 and Myo1c which is dynamic and actin dependent. Unlike wild-type Myo1c, the membrane localization of Neph1 was significantly reduced in podocytes expressing dominant negative Myo1c. In addition, Neph1 failed to localize at the podocyte cell membrane and cell junctions in Myo1c-depleted podocytes. We further demonstrate that similarly to Neph1, Myo1c also binds nephrin and reduces its localization at the podocyte cell membrane. A functional analysis of Myo1c knockdown cells showed defects in cell migration, as determined by a wound assay. In addition, the ability to form tight junctions was impaired in Myo1c knockdown cells, as determined by transepithelial electric resistance (TER) and bovine serum albumin (BSA) permeability assays. These results identify a novel Myo1c-dependent molecular mechanism that mediates the dynamic organization of Neph1 and nephrin at the slit diaphragm and is critical for podocyte function.
Collapse
|
39
|
Israël A. The IKK complex, a central regulator of NF-kappaB activation. Cold Spring Harb Perspect Biol 2010; 2:a000158. [PMID: 20300203 DOI: 10.1101/cshperspect.a000158] [Citation(s) in RCA: 652] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The IKK kinase complex is the core element of the NF-kappaB cascade. It is essentially made of two kinases (IKKalpha and IKKbeta) and a regulatory subunit, NEMO/IKKgamma. Additional components may exist, transiently or permanently, but their characterization is still unsure. In addition, it has been shown that two separate NF-kappaB pathways exist, depending on the activating signal and the cell type, the canonical (depending on IKKbeta and NEMO) and the noncanonical pathway (depending solely on IKKalpha). The main question, which is still only partially answered, is to understand how an NF-kappaB activating signal leads to the activation of the kinase subunits, allowing them to phosphorylate their targets and eventually induce nuclear translocation of the NF-kappaB dimers. I will review here the genetic, biochemical, and structural data accumulated during the last 10 yr regarding the function of the three IKK subunits.
Collapse
Affiliation(s)
- Alain Israël
- Unite de Signalisation Moleculaire et Activation Cellulaire, URA 2582 CNRS, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris Cedex 15, France.
| |
Collapse
|
40
|
McConnell RE, Tyska MJ. Leveraging the membrane - cytoskeleton interface with myosin-1. Trends Cell Biol 2010; 20:418-26. [PMID: 20471271 PMCID: PMC2897960 DOI: 10.1016/j.tcb.2010.04.004] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 04/15/2010] [Accepted: 04/16/2010] [Indexed: 12/19/2022]
Abstract
Class 1 myosins are small motor proteins with the ability to simultaneously bind to actin filaments and cellular membranes. Given their ability to generate mechanical force, and their high prevalence in many cell types, these molecules are well positioned to carry out several important biological functions at the interface of membrane and the actin cytoskeleton. Indeed, recent studies implicate these motors in endocytosis, exocytosis, release of extracellular vesicles, and the regulation of tension between membrane and the cytoskeleton. Many class 1 myosins also exhibit a load-dependent mechano-chemical cycle that enables them to maintain tension for long periods of time without hydrolyzing ATP. These properties put myosins-1 in a unique position to regulate dynamic membrane-cytoskeleton interactions and respond to physical forces during these events.
Collapse
Affiliation(s)
- Russell E. McConnell
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37205; USA
| | - Matthew J. Tyska
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37205; USA
| |
Collapse
|
41
|
Tanaka Y, Yujiri T, Tanaka M, Mitani N, Tanimura A, Tanizawa Y. Alteration of adipokines during peripheral blood stem cell mobilization induced by granulocyte colony-stimulating factor. J Clin Apher 2009; 24:205-8. [PMID: 19816958 DOI: 10.1002/jca.20212] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Adipokines, soluble mediators produced by adipocytes, have been shown to play a role in various physiological and pathological conditions. We investigated the involvement of adipokines in granulocyte colony-stimulating factor (G-CSF)-induced mobilization of hematopoietic stem cells in 21 healthy donors. We found that serum visfatin and resistin levels, but not leptin and adiponectin levels, were significantly elevated by G-CSF treatment. G-CSF treatment activated signaling proteins like extracellular signal-regulated kinase and stimulated secretion of visfatin from 3T3-L1 adipocytes. These findings suggest that some adipokines may play a role in G-CSF-induced mobilization of stem cells from the bone marrow into systemic circulation.
Collapse
Affiliation(s)
- Yoshinori Tanaka
- Department of Bio-Signal Analysis, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | | | | | | | | | | |
Collapse
|
42
|
McKenna JMD, Ostap EM. Kinetics of the interaction of myo1c with phosphoinositides. J Biol Chem 2009; 284:28650-9. [PMID: 19706607 DOI: 10.1074/jbc.m109.049791] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
myo1c is a single-headed myosin that dynamically links membranes to the actin cytoskeleton. A putative pleckstrin homology domain has been identified in the myo1c tail that binds phosphoinositides and soluble inositol phosphates with high affinity. However, the kinetics of association and dissociation and the influence of phospholipid composition on the kinetics have not been determined. Stopped-flow spectroscopy was used to measure the binding and dissociation of a recombinant myo1c construct containing the tail and regulatory domains (myo1c(IQ-tail)) to and from 100-nm diameter large unilamellar vesicles (LUVs). We found the time course of association of myo1c(IQ-tail) with LUVs containing 2% phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P(2)) followed a two-exponential time course, and the rate of the predominant fast phase depended linearly upon the total lipid concentration. The apparent second-order rate constant was approximately diffusion-limited. Increasing the molar ratio of anionic phospholipid by adding phosphatidylserine, additional PtdIns(4,5)P(2), or by situating PtdIns(4,5)P(2) in a more physiologically relevant lipid background increased the apparent association rate constant less than 2-fold. myo1c(IQ-tail) dissociated from PtdIns(4,5)P(2) at a slower rate (2.0 s(-1)) than the pleckstrin homology domain of phospholipase C-delta (13 s(-1)). The presence of additional anionic phospholipid reduced the myo1c(IQ-tail) dissociation rate constant >50-fold but marginally changed the dissociation rate of phospholipase C-delta, suggesting that additional electrostatic interactions in myo1c(IQ-tail) help to stabilize binding. Remarkably, high concentrations of soluble inositol phosphates induce dissociation of myo1c(IQ-tail) from LUVs, suggesting that phosphoinositides are able to bind to and dissociate from myo1c(IQ-tail) as it remains bound to the membrane.
Collapse
Affiliation(s)
- Jennine M Dawicki McKenna
- Department of Physiology, Pennsylvania Muscle Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6085, USA
| | | |
Collapse
|
43
|
Chariot A. The NF-kappaB-independent functions of IKK subunits in immunity and cancer. Trends Cell Biol 2009; 19:404-13. [PMID: 19648011 DOI: 10.1016/j.tcb.2009.05.006] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Revised: 05/13/2009] [Accepted: 05/19/2009] [Indexed: 12/14/2022]
Abstract
The I kappaB kinase (IKK) complex is involved in transcriptional activation by phosphorylating the inhibitory molecule I kappaB alpha, a modification that triggers its subsequent degradation, enabling activation of nuclear factor kappa B (NF-kappaB). Importantly, recent reports indicate that multiple cytoplasmic and nuclear proteins distinct from the NF-kappaB and I kappaB proteins are phosphorylated by the catalytic subunits of the IKK complex, IKK alpha or IKK beta. Here, I describe how IKK subunits can have crucial roles in allergy, inflammation and immunity by targeting proteins such as SNAP23 and IRF7, but also in cancer by phosphorylating key molecules such as p53, TSC1 and FOXO3a through NF-kappaB-independent pathways. Thus, these recent findings considerably widen the biological roles of these kinases and suggest that a full understanding of the biological roles of IKK alpha and IKK beta requires an exhaustive characterization of their substrates.
Collapse
Affiliation(s)
- Alain Chariot
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), GIGA-Signal Transduction, Laboratory of Medical Chemistry, CHU, Sart-Tilman, University of Liege, Belgium.
| |
Collapse
|
44
|
Abstract
Insulin signaling at target tissues is essential for growth and development and for normal homeostasis of glucose, fat, and protein metabolism. Control over this process is therefore tightly regulated. It can be achieved by a negative feedback control mechanism whereby downstream components inhibit upstream elements along the insulin-signaling pathway (autoregulation) or by signals from apparently unrelated pathways that inhibit insulin signaling thus leading to insulin resistance. Phosphorylation of insulin receptor substrate (IRS) proteins on serine residues has emerged as a key step in these control processes under both physiological and pathological conditions. The list of IRS kinases implicated in the development of insulin resistance is growing rapidly, concomitant with the list of potential Ser/Thr phosphorylation sites in IRS proteins. Here, we review a range of conditions that activate IRS kinases to phosphorylate IRS proteins on "hot spot" domains. The flexibility vs. specificity features of this reaction is discussed and its characteristic as an "array" phosphorylation is suggested. Finally, its implications on insulin signaling, insulin resistance and type 2 diabetes, an emerging epidemic of the 21st century are outlined.
Collapse
Affiliation(s)
- Sigalit Boura-Halfon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 76100 Israel
| | | |
Collapse
|
45
|
Boura-Halfon S, Zick Y. Serine kinases of insulin receptor substrate proteins. VITAMINS AND HORMONES 2009; 80:313-49. [PMID: 19251043 DOI: 10.1016/s0083-6729(08)00612-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Signaling of insulin and insulin-like growth factor-I (IGF-1) at target tissues is essential for growth, development and for normal homeostasis of glucose, fat, and protein metabolism. Control over this process is therefore tightly regulated. It can be achieved by a negative-feedback control mechanism, whereby downstream components inhibit upstream elements along the insulin and IGF-1 signaling pathway or by signals from other pathways that inhibit insulin/IGF-1 signaling thus leading to insulin/IGF-1 resistance. Phosphorylation of insulin receptor substrates (IRS) proteins on serine residues has emerged as a key step in these control processes both under physiological and pathological conditions. The list of IRS kinases is growing rapidly, concomitant with the list of potential Ser/Thr phosphorylation sites in IRS proteins. Here we review a range of conditions that activate IRS kinases to phosphorylate IRS proteins on selected domains. The specificity of this reaction is discussed and its characteristic as an "array" phosphorylation is suggested. Finally, its implications on insulin/IGF-1 signaling, insulin/IGF-1 resistance and diabetes, an emerging epidemic of the twenty-first century are outlined.
Collapse
Affiliation(s)
- Sigalit Boura-Halfon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | |
Collapse
|
46
|
GβL regulates TNFα-induced NF-кB signaling by directly inhibiting the activation of IкB kinase. Cell Signal 2008; 20:2127-33. [DOI: 10.1016/j.cellsig.2008.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Accepted: 08/04/2008] [Indexed: 11/19/2022]
|
47
|
Kaddai V, Gonzalez T, Bolla M, Le Marchand-Brustel Y, Cormont M. The nitric oxide-donating derivative of acetylsalicylic acid, NCX 4016, stimulates glucose transport and glucose transporters translocation in 3T3-L1 adipocytes. Am J Physiol Endocrinol Metab 2008; 295:E162-9. [PMID: 18492771 DOI: 10.1152/ajpendo.00622.2007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
NCX 4016 is a nitric oxide (NO)-donating derivative of acetylsalicylic acid. NO and salicylate, in vivo metabolites of NCX 4016, were shown to be potential actors in controlling glucose homeostasis. In this study, we evaluated the action of NCX 4016 on the capacity of 3T3-L1 adipocytes to transport glucose in basal and insulin-stimulated conditions. NCX 4016 induced a twofold increase in glucose uptake in parallel with the translocation of the glucose transporters GLUT1 and GLUT4 to the plasma membrane, leaving unaffected their total expression levels. Importantly, NCX 4016 further increased glucose transport induced by a physiological concentration of insulin. The stimulatory effect of NCX 4016 on glucose uptake appears to be mediated by its NO moiety. Indeed, it is inhibited by a NO scavenger and treatment with acetylsalicylic or salicylic acid had no effect. Although NO is involved in the action of NCX 4016, it did not mainly depend on the soluble cGMP cyclase/protein kinase G pathway. Furthermore, NCX 4016-stimulated glucose transport did not involve the insulin-signaling cascade required to stimulate glucose transport. NCX 4016 induces a small activation of the mitogen-activated protein kinases p38 and c-Jun NH(2)-terminal kinase and no activation of other stress-activated signaling molecules, including extracellular signal-regulated kinase, inhibitory factor kappaB, or AMP-activated kinases. Interestingly, NCX 4016 modified the content of S-nitrosylated proteins in adipocytes. Taken together, our results indicate that NCX 4016 induced glucose transport in adipocytes through a novel mechanism possibly involving S-nitrosylation. NCX 4016 thus possesses interesting characteristics to be considered as a candidate molecule for the treatment of patients suffering from metabolic syndrome and type 2 diabetes.
Collapse
Affiliation(s)
- V Kaddai
- Institut National de la Santé et de la Recherche Médicale Unité 895, Cellular and Molecular Physiopathology of Obesity and Diabetes, Faculté de Médecine, University of Nice/Sophia-Antipolis, Nice, France
| | | | | | | | | |
Collapse
|
48
|
Roher N, Samokhvalov V, Díaz M, MacKenzie S, Klip A, Planas JV. The proinflammatory cytokine tumor necrosis factor-alpha increases the amount of glucose transporter-4 at the surface of muscle cells independently of changes in interleukin-6. Endocrinology 2008; 149:1880-9. [PMID: 18162526 DOI: 10.1210/en.2007-1045] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
TNFalpha is a proinflammatory cytokine secreted by macrophages in response to bacterial infection. Recently new evidence has emerged suggesting that stressed or injured myocytes produce TNFalpha that then acts as an autocrine and/or paracrine mediator. TNFalpha receptors types 1 and 2 are present in skeletal muscle cells, and muscle cells can secrete, in addition to TNFalpha, other cytokines such as IL-1beta or IL-6. Furthermore, the plasma concentration of TNFalpha is elevated in insulin-resistant states associated with obesity and type 2 diabetes. Here we show that TNFalpha increased the amount of glucose transporter (GLUT)-4 at the plasma membrane and also glucose uptake in the L6 muscle cell line stably expressing GLUT4 tagged with the c-myc epitope. Regardless of the state of differentiation of the L6 cells, TNFalpha did not affect the rate of proliferation or of apoptosis. The stimulatory effects of TNFalpha on cell surface GLUT4 and glucose uptake were blocked by nuclear factor-kappaB and p38MAPK pathway specific inhibitors (Bay 11-7082 and SB220025), and these two pathways were stimulated by TNFalpha. Furthermore, although TNFalpha increased IL-6 mRNA and protein expression, IL-6 did not mediate the effects of TNFalpha on cell surface GLUT4 levels, which also did not require de novo protein synthesis. The results indicate that TNFalpha can stimulate glucose uptake in L6 muscle cells by inducing GLUT4 translocation to the plasma membrane, possibly through activation of the nuclear factor-kappaB and p38MAPK signaling pathways and independently of the production of IL-6.
Collapse
Affiliation(s)
- Nerea Roher
- Departament de Fisiologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
49
|
Sheng CH, Di J, Jin Y, Zhang YC, Wu M, Sun Y, Zhang GZ. Resistin is expressed in human hepatocytes and induces insulin resistance. Endocrine 2008; 33:135-43. [PMID: 18446452 DOI: 10.1007/s12020-008-9065-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 03/27/2008] [Accepted: 04/14/2008] [Indexed: 02/06/2023]
Abstract
Resistin, known as an adipocyte-specific secretory factor (ADSF), is implicated to modulate insulin resistance in rodents. However, the precise role of this factor for human insulin resistance has remained elusive. Here, we investigate the relationship between human resistin and insulin resistance in hepatocytes and the effect of Metformin on resistin. In this study, the expression of resistin in human hepatocytes and hepatic tissues was examined, and the human resistin eukaryotic expression vector was constructed and stably transfected in HepG2 cells. Data showed that resistin is expressed in human hepatocytes and hepatic tissues. Overexpression of human resistin impaired significantly insulin-stimulated glucose uptake and glycogen synthesis in HepG2 cells. It also decreased the expression of insulin receptor substrate 2 (IRS-2) and c-cbl associated protein (CAP), whereas increased the expression of glycogen synthetase kinase 3beta (GSK-3beta). The result suggested that human resistin induced insulin resistance in hepatocytes by blocking the two insulin signal transduction pathways of PI-3K/Akt and of CAP/c-cbl. We also concluded that Metformin reversed the effect of resistin and downregulated the expression of resistin in hepatocytes.
Collapse
Affiliation(s)
- Chun Hua Sheng
- Department of Central Research, The Third Clinical College, Jilin University, Xiantai Street No. 126, Changchun, 130033, China
| | | | | | | | | | | | | |
Collapse
|
50
|
Yoshida T, Yamagishi S, Matsui T, Nakamura K, Ueno T, Takeuchi M, Sata M. Telmisartan, an Angiotensin II Type 1 Receptor Blocker, Inhibits Advanced Glycation End-product (AGE)-elicited Hepatic Insulin Resistance via Peroxisome Proliferator-activated Receptor-γ Activation. J Int Med Res 2008; 36:237-43. [DOI: 10.1177/147323000803600204] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
This study examined whether telmisartan, a unique angiotensin II type 1 receptor blocker (ARB) with peroxisome proliferator-activated receptor-γ (PPAR-γ)-modulating activity, improved insulin resistance in advanced glycation end-product (AGE)-exposed human hepatoma (Hep3B) cells. AGE increased phosphorylation of insulin receptor substrate-1 (IRS-1) at serine-307 residues in Hep3B cells. It also decreased tyrosine phosphorylation of IRS-1 and, subsequently, reduced the association of the p85 subunit of phosphatidylinositol 3-kinase with IRS-1 and glycogen synthesis in insulin-exposed Hep3B cells, all of which were inhibited by telmisartan. The insulin-sensitizing properties of telmisartan in AGE-exposed Hep3B cells were significantly blocked by GW9662, an inhibitor of PPAR-γ. Candesartan, another ARB, did not affect AGEs-induced serine phosphorylation of IRS-1 at serine-307 residues in Hep3B cells. Our study suggests that telmisartan could improve AGE-elicited insulin resistance in Hep3B cells by inhibiting serine phosphorylation of IRS-1, at least in part, via activation of PPAR-γ. Telmisartan may play a protective role against hepatic insulin resistance in diabetes.
Collapse
Affiliation(s)
- T Yoshida
- Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - S Yamagishi
- Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - T Matsui
- Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - K Nakamura
- Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - T Ueno
- Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - M Takeuchi
- Department of Pathophysiological Science, Faculty of Pharmaceutical Science, Hokuriku University, Kanazawa, Japan
| | - M Sata
- Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|