1
|
Maharjan S, Kirk RS, Lawton SP, Walker AJ. Human growth factor-mediated signalling through lipid rafts regulates stem cell proliferation, development and survival of Schistosoma mansoni. Open Biol 2024; 14:230262. [PMID: 38195062 PMCID: PMC10776228 DOI: 10.1098/rsob.230262] [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: 08/03/2023] [Accepted: 11/16/2023] [Indexed: 01/11/2024] Open
Abstract
Although the mechanisms by which schistosomes grow and develop in humans are poorly defined, their unique outer tegument layer, which interfaces with host blood, is considered vital to homeostasis of the parasite. Here, we investigated the importance of tegument lipid rafts to the biology of Schistosoma mansoni in the context of host-parasite interactions. We demonstrate the temporal clustering of lipid rafts in response to human epidermal growth factor (EGF) during early somule development, concomitant with the localization of anteriorly orientated EGF receptors (EGFRs) and insulin receptors, mapped using fluorescent EGF/insulin ligand. Methyl-β-cyclodextrin (MβCD)-mediated depletion of cholesterol from lipid rafts abrogated the EGFR/IR binding at the parasite surface and led to modulation of protein kinase C, extracellular signal-regulated kinase, p38 mitogen-activated protein kinase and Akt signalling pathways within the parasite. Furthermore, MβCD-mediated lipid raft disruption, and blockade of EGFRs using canertinib, profoundly reduced somule motility and survival, and attenuated stem cell proliferation and somule growth and development particularly to the fast-growing liver stage. These findings provide a novel paradigm for schistosome development and vitality in the host, driven through host-parasite interactions at the tegument, that might be exploitable for developing innovative therapeutic approaches to combat human schistosomiasis.
Collapse
Affiliation(s)
- Shradha Maharjan
- Molecular Parasitology Laboratory, School of Life Sciences, Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames KT1 2EE, UK
| | - Ruth S. Kirk
- Molecular Parasitology Laboratory, School of Life Sciences, Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames KT1 2EE, UK
| | - Scott P. Lawton
- Centre for Epidemiology and Planetary Health, SRUC School of Veterinary Medicine, Scotland's Rural College, West Mains Road, Edinburgh EH9 3JG, UK
| | - Anthony J. Walker
- Molecular Parasitology Laboratory, School of Life Sciences, Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames KT1 2EE, UK
| |
Collapse
|
2
|
Salvador CL, Oppebøen M, Vassli AØ, Pfeiffer HCV, Varhaug KN, Elgstøen KBP, Yazdani M. Increased Sphingomyelin and Free Sialic Acid in Cerebrospinal Fluid of Kearns-Sayre Syndrome: New Findings Using Untargeted Metabolomics. Pediatr Neurol 2023; 143:68-76. [PMID: 37018879 DOI: 10.1016/j.pediatrneurol.2023.02.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/10/2023] [Accepted: 02/25/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Kearns-Sayre syndrome (KSS) is caused by duplications and/or deletions of mitochondrial DNA (mtDNA) and is typically diagnosed based on a classic triad of symptoms with chronic progressive external ophthalmoplegia (CPEO), retinitis pigmentosa, and onset before age 20 years. The present study aimed to diagnose two patients, on suspicion of KSS. METHODS One of the patients went through a diagnostic odyssey, with normal results from several mtDNA analyses, both in blood and muscle, before the diagnosis was confirmed genetically. RESULTS Two patients presented increased tau protein and low 5-methyltetrahydrofolate (5-MTHF) levels in the cerebrospinal fluid (CSF). Untargeted metabolomics on CSF samples also showed an increase in the levels of free sialic acid and sphingomyelin C16:0 (d18:1/C16:0), compared with four control groups (patients with mitochondrial disorders, nonmitochondrial disorders, low 5-MTHF, or increased tau proteins). CONCLUSIONS It is the first time that elevated sphingomyelin C16:0 (d18:1/C16:0) and tau protein in KSS are reported. Using an untargeted metabolomics approach and standard laboratory methods, the study could shed new light on metabolism in KSS to better understand its complexity. In addition, the findings may suggest the combination of elevated free sialic acid, sphingomyelin C16:0 (d18:1/C16:0), and tau protein as well as low 5-MTHF as new biomarkers in the diagnostics of KSS.
Collapse
Affiliation(s)
| | - Mari Oppebøen
- Department of Pediatrics, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Anja Østeby Vassli
- Department of Medical Biochemistry, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Helle Cecilie Viekilde Pfeiffer
- Department of Pediatrics, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Department of Pediatrics, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Kristin Nielsen Varhaug
- The Mitochondrial Medicine and Neurogenetics (MMN) Group, Department of Clinical Medicine, University of Bergen, Bergen, Norway; Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | | | - Mazyar Yazdani
- Department of Medical Biochemistry, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
3
|
Tian Y, Mehta K, Jellinek MJ, Sun H, Lu W, Shi R, Ingram K, Friedline RH, Kim JK, Kemper JK, Ford DA, Zhang K, Wang B. Hepatic Phospholipid Remodeling Modulates Insulin Sensitivity and Systemic Metabolism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300416. [PMID: 37088778 PMCID: PMC10288282 DOI: 10.1002/advs.202300416] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/13/2023] [Indexed: 05/03/2023]
Abstract
The liver plays a central role in regulating glucose and lipid metabolism. Aberrant insulin action in the liver is a major driver of selective insulin resistance, in which insulin fails to suppress glucose production but continues to activate lipogenesis in the liver, resulting in hyperglycemia and hypertriglyceridemia. The underlying mechanisms of selective insulin resistance are not fully understood. Here It is shown that hepatic membrane phospholipid composition controlled by lysophosphatidylcholine acyltransferase 3 (LPCAT3) regulates insulin signaling and systemic glucose and lipid metabolism. Hyperinsulinemia induced by high-fat diet (HFD) feeding augments hepatic Lpcat3 expression and membrane unsaturation. Loss of Lpcat3 in the liver improves insulin resistance and blunts lipogenesis in both HFD-fed and genetic ob/ob mouse models. Mechanistically, Lpcat3 deficiency directly facilitates insulin receptor endocytosis, signal transduction, and hepatic glucose production suppression and indirectly enhances fibroblast growth factor 21 (FGF21) secretion, energy expenditure, and glucose uptake in adipose tissue. These findings identify hepatic LPCAT3 and membrane phospholipid composition as a novel regulator of insulin sensitivity and provide insights into the pathogenesis of selective insulin resistance.
Collapse
Affiliation(s)
- Ye Tian
- Department of Comparative BiosciencesCollege of Veterinary MedicineUniversity of Illinois at Urbana‐ChampaignUrbanaIL61802USA
| | - Kritika Mehta
- Department of BiochemistrySchool of Molecular and Cellular BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Matthew J. Jellinek
- Department of Biochemistry and Molecular Biologyand Center for Cardiovascular ResearchSaint Louis UniversitySt. LouisMO63104USA
| | - Hao Sun
- Department of Molecular and Integrative PhysiologySchool of Molecular and Cellular BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Wei Lu
- Department of Comparative BiosciencesCollege of Veterinary MedicineUniversity of Illinois at Urbana‐ChampaignUrbanaIL61802USA
| | - Ruicheng Shi
- Department of Comparative BiosciencesCollege of Veterinary MedicineUniversity of Illinois at Urbana‐ChampaignUrbanaIL61802USA
| | - Kevin Ingram
- Department of BiochemistrySchool of Molecular and Cellular BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Randall H. Friedline
- Program in Molecular Medicine and Division of EndocrinologyMetabolism and DiabetesDepartment of MedicineUniversity of Massachusetts Medical SchoolWorcesterMA01655USA
| | - Jason K. Kim
- Program in Molecular Medicine and Division of EndocrinologyMetabolism and DiabetesDepartment of MedicineUniversity of Massachusetts Medical SchoolWorcesterMA01655USA
| | - Jongsook Kim Kemper
- Department of Molecular and Integrative PhysiologySchool of Molecular and Cellular BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
- Cancer Center at IllinoisUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - David A. Ford
- Department of Biochemistry and Molecular Biologyand Center for Cardiovascular ResearchSaint Louis UniversitySt. LouisMO63104USA
| | - Kai Zhang
- Department of BiochemistrySchool of Molecular and Cellular BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Bo Wang
- Department of Comparative BiosciencesCollege of Veterinary MedicineUniversity of Illinois at Urbana‐ChampaignUrbanaIL61802USA
- Cancer Center at IllinoisUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
- Division of Nutritional SciencesCollege of AgriculturalConsumer and Environmental SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| |
Collapse
|
4
|
Wang L, Wiedmann TS, Kandimalla KK. Modulating insulin signaling and trafficking at the blood-brain barrier endothelium using lipid based nanoemulsions. Int J Pharm 2022; 622:121823. [PMID: 35605891 PMCID: PMC9881744 DOI: 10.1016/j.ijpharm.2022.121823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 05/01/2022] [Accepted: 05/08/2022] [Indexed: 01/31/2023]
Abstract
The compositionally distinct lipid rafts present in the plasma membrane regulate the restrictive trafficking and signal transduction in the blood-brain barrier (BBB) endothelium. Several metabolic and neurodegenerative diseases are associated with lipid homeostasis disruption within the BBB endothelium. Here, we hypothesized that the delivery of lipid triglyceride based nanoemulsions containing unsaturated fatty acids (UFAs) provides a novel non-pharmacological approach to modulate lipid raft integrity and rectify the aberrant trafficking and signal transduction. The current study has shown that soybean oil nanoemulsions (SNEs) altered the morphology of lipid rafts that are stained by Alex Fluor 647 labelled cholera toxin (AF647-CTX) in polarized human cerebral microvascular endothelial (hCMEC/D3) cell monolayers. Moreover, western blot and flow cytometry analysis showed that SNEs containing polyunsaturated fatty acids (PUFAs) increased phospo-AKT (p-AKT) expression, a marker for the stimulation of metabolic arm of insulin signaling, and insulin uptake in hCMEC/D3 monolayers. However, olive oil nanoemulsions (ONEs) containing monounsaturated fatty acids (MUFAs) had no detectable impact on lipid raft integrity, AKT phosphorylation, or insulin uptake. These findings provided direct evidence that SNEs containing PUFAs can upregulate insulin-pAKT pathway, facilitate insulin trafficking at the BBB, and potentially address cerebrovascular dysfunction in metabolic and neurodegenerative diseases.
Collapse
Affiliation(s)
- Lushan Wang
- Department of Pharmaceutics, University of Minnesota, College of Pharmacy, Minneapolis, MN 55455, United States,Brain Barriers Research Center, University of Minnesota, College of Pharmacy, Minneapolis, MN 55455, United States
| | - Timothy S. Wiedmann
- Department of Pharmaceutics, University of Minnesota, College of Pharmacy, Minneapolis, MN 55455, United States
| | - Karunya K. Kandimalla
- Department of Pharmaceutics, University of Minnesota, College of Pharmacy, Minneapolis, MN 55455, United States,Brain Barriers Research Center, University of Minnesota, College of Pharmacy, Minneapolis, MN 55455, United States,Corresponding author. (K.K. Kandimalla)
| |
Collapse
|
5
|
Deep proteomic profiling unveils arylsulfatase A as a non-alcoholic steatohepatitis inducible hepatokine and regulator of glycemic control. Nat Commun 2022; 13:1259. [PMID: 35273160 PMCID: PMC8913628 DOI: 10.1038/s41467-022-28889-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 02/10/2022] [Indexed: 12/13/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) and type 2 diabetes are closely linked, yet the pathophysiological mechanisms underpinning this bidirectional relationship remain unresolved. Using proteomic approaches, we interrogate hepatocyte protein secretion in two models of murine NASH to understand how liver-derived factors modulate lipid metabolism and insulin sensitivity in peripheral tissues. We reveal striking hepatokine remodelling that is associated with insulin resistance and maladaptive lipid metabolism, and identify arylsulfatase A (ARSA) as a hepatokine that is upregulated in NASH and type 2 diabetes. Mechanistically, hepatic ARSA reduces sulfatide content and increases lysophosphatidylcholine (LPC) accumulation within lipid rafts and suppresses LPC secretion from the liver, thereby lowering circulating LPC and lysophosphatidic acid (LPA) levels. Reduced LPA is linked to improvements in skeletal muscle insulin sensitivity and systemic glycemic control. Hepatic silencing of Arsa or inactivation of ARSA's enzymatic activity reverses these effects. Together, this study provides a unique resource describing global changes in hepatokine secretion in NASH, and identifies ARSA as a regulator of liver to muscle communication and as a potential therapeutic target for type 2 diabetes.
Collapse
|
6
|
Corrêa T, Feltes BC, Giugliani R, Matte U. Disruption of morphogenic and growth pathways in lysosomal storage diseases. WIREs Mech Dis 2021; 13:e1521. [PMID: 34730292 DOI: 10.1002/wsbm.1521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/12/2020] [Accepted: 01/21/2021] [Indexed: 12/11/2022]
Abstract
The lysosome achieved a new protagonism that highlights its multiple cellular functions, such as in the catabolism of complex substrates, nutrient sensing, and signaling pathways implicated in cell metabolism and growth. Lysosomal storage diseases (LSDs) cause lysosomal accumulation of substrates and deficiency in trafficking of macromolecules. The substrate accumulation can impact one or several pathways which contribute to cell damage. Autophagy impairment and immune response are widely studied, but less attention is paid to morphogenic and growth pathways and its impact on the pathophysiology of LSDs. Hedgehog pathway is affected with abnormal expression and changes in distribution of protein levels, and a reduced number and length of primary cilia. Moreover, growth pathways are identified with delay in reactivation of mTOR that deregulate termination of autophagy and reformation of lysosomes. Insulin resistance caused by changes in lipids rafts has been described in different LSDs. While the genetic and biochemical bases of deficient proteins in LSDs are well understood, the secondary molecular mechanisms that disrupt wider biological processes associated with LSDs are only now becoming clearer. Therefore, we explored how specific signaling pathways can be related to specific LSDs, showing that a system medicine approach could be a valuable tool for the better understanding of LSD pathogenesis. This article is categorized under: Metabolic Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Thiago Corrêa
- Department of Genetics, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Bruno C Feltes
- Department of Theoretical Informatics, Institute of Informatics, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Roberto Giugliani
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Ursula Matte
- Department of Genetics, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
7
|
Suresh P, Miller WT, London E. Phospholipid exchange shows insulin receptor activity is supported by both the propensity to form wide bilayers and ordered raft domains. J Biol Chem 2021; 297:101010. [PMID: 34324831 PMCID: PMC8379460 DOI: 10.1016/j.jbc.2021.101010] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 11/29/2022] Open
Abstract
Insulin receptor (IR) is a membrane tyrosine kinase that mediates the response of cells to insulin. IR activity has been shown to be modulated by changes in plasma membrane lipid composition, but the properties and structural determinants of lipids mediating IR activity are poorly understood. Here, using efficient methyl-alpha-cyclodextrin mediated lipid exchange, we studied the effect of altering plasma membrane outer leaflet phospholipid composition upon the activity of IR in mammalian cells. After substitution of endogenous lipids with lipids having an ability to form liquid ordered (Lo) domains (sphingomyelins) or liquid disordered (Ld) domains (unsaturated phosphatidylcholines (PCs)), we found that the propensity of lipids to form ordered domains is required for high IR activity. Additional substitution experiments using a series of saturated PCs showed that IR activity increased substantially with increasing acyl chain length, which increases both bilayer width and the propensity to form ordered domains. Incorporating purified IR into alkyl maltoside micelles with increasing hydrocarbon lengths also increased IR activity, but more modestly than by increasing lipid acyl chain length in cells. These results suggest that the ability to form Lo domains as well as wide bilayer width contributes to increased IR activity. Inhibition of phosphatases showed that some of the lipid dependence of IR activity upon lipid structure reflected protection from phosphatases by lipids that support Lo domain formation. These results are consistent with a model in which a combination of bilayer width and ordered domain formation modulates IR activity via IR conformation and accessibility to phosphatases.
Collapse
Affiliation(s)
- Pavana Suresh
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, USA
| | - W Todd Miller
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, New York, USA; Department of Veterans Affairs Medical Center, Northport, New York, USA
| | - Erwin London
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, USA.
| |
Collapse
|
8
|
Lair B, Laurens C, Van Den Bosch B, Moro C. Novel Insights and Mechanisms of Lipotoxicity-Driven Insulin Resistance. Int J Mol Sci 2020; 21:E6358. [PMID: 32887221 PMCID: PMC7504171 DOI: 10.3390/ijms21176358] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/27/2020] [Accepted: 08/27/2020] [Indexed: 02/07/2023] Open
Abstract
A large number of studies reported an association between elevated circulating and tissue lipid content and metabolic disorders in obesity, type 2 diabetes (T2D) and aging. This state of uncontrolled tissue lipid accumulation has been called lipotoxicity. It was later shown that excess lipid flux is mainly neutralized within lipid droplets as triglycerides, while several bioactive lipid species such as diacylglycerols (DAGs), ceramides and their derivatives have been mechanistically linked to the pathogenesis of insulin resistance (IR) by antagonizing insulin signaling and action in metabolic organs such as the liver and skeletal muscle. Skeletal muscle and the liver are the main sites of glucose disposal in the body and IR in these tissues plays a pivotal role in the development of T2D. In this review, we critically examine recent literature supporting a causal role of DAGs and ceramides in the development of IR. A particular emphasis is placed on transgenic mouse models with modulation of total DAG and ceramide pools, as well as on modulation of specific subspecies, in relation to insulin sensitivity. Collectively, although a wide number of studies converge towards the conclusion that both DAGs and ceramides cause IR in metabolic organs, there are still some uncertainties on their mechanisms of action. Recent studies reveal that subcellular localization and acyl chain composition are determinants in the biological activity of these lipotoxic lipids and should be further examined.
Collapse
Affiliation(s)
- Benjamin Lair
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; (B.L.); (C.L.); (B.V.D.B.)
- University of Toulouse, Paul Sabatier University, 31330 Toulouse, France
| | - Claire Laurens
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; (B.L.); (C.L.); (B.V.D.B.)
- University of Toulouse, Paul Sabatier University, 31330 Toulouse, France
| | - Bram Van Den Bosch
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; (B.L.); (C.L.); (B.V.D.B.)
- University of Toulouse, Paul Sabatier University, 31330 Toulouse, France
| | - Cedric Moro
- INSERM, UMR1048, Institute of Metabolic and Cardiovascular Diseases, 31432 Toulouse, France; (B.L.); (C.L.); (B.V.D.B.)
- University of Toulouse, Paul Sabatier University, 31330 Toulouse, France
| |
Collapse
|
9
|
Torres M, Rosselló CA, Fernández-García P, Lladó V, Kakhlon O, Escribá PV. The Implications for Cells of the Lipid Switches Driven by Protein-Membrane Interactions and the Development of Membrane Lipid Therapy. Int J Mol Sci 2020; 21:ijms21072322. [PMID: 32230887 PMCID: PMC7177374 DOI: 10.3390/ijms21072322] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/17/2020] [Accepted: 03/19/2020] [Indexed: 02/06/2023] Open
Abstract
The cell membrane contains a variety of receptors that interact with signaling molecules. However, agonist-receptor interactions not always activate a signaling cascade. Amphitropic membrane proteins are required for signal propagation upon ligand-induced receptor activation. These proteins localize to the plasma membrane or internal compartments; however, they are only activated by ligand-receptor complexes when both come into physical contact in membranes. These interactions enable signal propagation. Thus, signals may not propagate into the cell if peripheral proteins do not co-localize with receptors even in the presence of messengers. As the translocation of an amphitropic protein greatly depends on the membrane's lipid composition, regulation of the lipid bilayer emerges as a novel therapeutic strategy. Some of the signals controlled by proteins non-permanently bound to membranes produce dramatic changes in the cell's physiology. Indeed, changes in membrane lipids induce translocation of dozens of peripheral signaling proteins from or to the plasma membrane, which controls how cells behave. We called these changes "lipid switches", as they alter the cell's status (e.g., proliferation, differentiation, death, etc.) in response to the modulation of membrane lipids. Indeed, this discovery enables therapeutic interventions that modify the bilayer's lipids, an approach known as membrane-lipid therapy (MLT) or melitherapy.
Collapse
Affiliation(s)
- Manuel Torres
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Ctra. de Valldemossa km 7.5, E-07122 Palma, Spain; (M.T.); (C.A.R.); (P.F.-G.); (V.L.)
- Department of R&D, Laminar Pharmaceuticals SL. ParcBit, Ed. Naorte B, E-07121 Palma, Spain
| | - Catalina Ana Rosselló
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Ctra. de Valldemossa km 7.5, E-07122 Palma, Spain; (M.T.); (C.A.R.); (P.F.-G.); (V.L.)
- Department of R&D, Laminar Pharmaceuticals SL. ParcBit, Ed. Naorte B, E-07121 Palma, Spain
| | - Paula Fernández-García
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Ctra. de Valldemossa km 7.5, E-07122 Palma, Spain; (M.T.); (C.A.R.); (P.F.-G.); (V.L.)
- Department of R&D, Laminar Pharmaceuticals SL. ParcBit, Ed. Naorte B, E-07121 Palma, Spain
| | - Victoria Lladó
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Ctra. de Valldemossa km 7.5, E-07122 Palma, Spain; (M.T.); (C.A.R.); (P.F.-G.); (V.L.)
- Department of R&D, Laminar Pharmaceuticals SL. ParcBit, Ed. Naorte B, E-07121 Palma, Spain
| | - Or Kakhlon
- Department of Neurology, Hadassah-Hebrew University Medical Center, Ein Kerem, 91120 Jerusalem, Israel;
| | - Pablo Vicente Escribá
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, Ctra. de Valldemossa km 7.5, E-07122 Palma, Spain; (M.T.); (C.A.R.); (P.F.-G.); (V.L.)
- Correspondence:
| |
Collapse
|
10
|
Kałużna M, Trzeciak I, Ziemnicka K, Machaczka M, Ruchała M. Endocrine and metabolic disorders in patients with Gaucher disease type 1: a review. Orphanet J Rare Dis 2019; 14:275. [PMID: 31791361 PMCID: PMC6889605 DOI: 10.1186/s13023-019-1211-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/24/2019] [Indexed: 12/26/2022] Open
Abstract
Background Gaucher disease (GD) is one of the most prevalent lysosomal storage diseases and is associated with hormonal and metabolic abnormalities, including nutritional status disorders, hypermetabolic state with high resting energy expenditures, peripheral insulin resistance, hypoadiponectinaemia, leptin and ghrelin impairments, hypolipidaemia, linear growth deceleration and growth hormone deficiency, delayed puberty, hypocalcaemia and vitamin D deficiency. Specific treatments for GD such as enzyme replacement therapy and substrate reduction therapy display significant effects on the metabolic profile of GD patients. Main body of the abstract Hormonal and metabolic disturbances observed in both adult and paediatric patients with Gaucher disease type 1 (GD1) are discussed in this review. The PubMed database was used to identify articles on endocrine and metabolic disorders in GD1. GD1 appears to facilitate the development of disorders of nutrition, glucose metabolism and vitamin D insufficiency. Metabolic and hormonal diseases may have a significant impact on the course of the underlying disease and patient quality of life. Conclusions Conditions relating to hormones and metabolism can be wide-ranging in GD1. Obtained findings were intrinsic to GD either as a deleterious process or a compensatory response and some changes detected may represent co-morbidities. Actively seeking and diagnosing endocrine and metabolic disorders are strongly recommended in GD1 patients to optimize healthcare.
Collapse
Affiliation(s)
- Małgorzata Kałużna
- Ward of Endocrinology, Metabolism and Internal Diseases Ward, Heliodor Swiecicki University Hospital, Poznan, Poland. .,Department of Endocrinology Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland.
| | - Isabella Trzeciak
- Department of Endocrinology Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Ziemnicka
- Ward of Endocrinology, Metabolism and Internal Diseases Ward, Heliodor Swiecicki University Hospital, Poznan, Poland.,Department of Endocrinology Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Machaczka
- Medical Faculty, University of Rzeszow, Rzeszow, Poland.,Department of Clinical Science and Education, Division of Internal Medicine, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Marek Ruchała
- Ward of Endocrinology, Metabolism and Internal Diseases Ward, Heliodor Swiecicki University Hospital, Poznan, Poland.,Department of Endocrinology Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
11
|
Huang Q, Kahn CR, Altindis E. Viral Hormones: Expanding Dimensions in Endocrinology. Endocrinology 2019; 160:2165-2179. [PMID: 31310273 PMCID: PMC6736053 DOI: 10.1210/en.2019-00271] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/10/2019] [Indexed: 02/07/2023]
Abstract
Viruses have developed different mechanisms to manipulate their hosts, including the process of viral mimicry in which viruses express important host proteins. Until recently, examples of viral mimicry were limited to mimics of growth factors and immunomodulatory proteins. Using a comprehensive bioinformatics approach, we have shown that viruses possess the DNA/RNA with potential to encode 16 different peptides with high sequence similarity to human peptide hormones and metabolically important regulatory proteins. We have characterized one of these families, the viral insulin/IGF-1-like peptides (VILPs), which we identified in four members of the Iridoviridae family. VILPs can bind to human insulin and IGF-1 receptors and stimulate classic postreceptor signaling pathways. Moreover, VILPs can stimulate glucose uptake in vitro and in vivo and stimulate DNA synthesis. DNA sequences of some VILP-carrying viruses have been identified in the human enteric virome. In addition to VILPs, sequences with homology to 15 other peptide hormones or cytokines can be identified in viral DNA/RNA sequences, some with a very high identity to hormones. Recent data by others has identified a peptide that resembles and mimics α-melanocyte-stimulating hormone's anti-inflammatory effects in in vitro and in vivo models. Taken together, these studies reveal novel mechanisms of viral and bacterial pathogenesis in which the microbe can directly target or mimic the host endocrine system. These findings also introduce the concept of a system of microbial hormones that provides new insights into the evolution of peptide hormones, as well as potential new roles of microbial hormones in health and disease.
Collapse
Affiliation(s)
- Qian Huang
- Boston College Biology Department, Chestnut Hill, Massachusetts
| | - C Ronald Kahn
- Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Emrah Altindis
- Boston College Biology Department, Chestnut Hill, Massachusetts
- Correspondence: Emrah Altindis, PhD, Boston College Biology Department, Higgins Hall 515, 140 Commonwealth Avenue, Chestnut Hill, Massachusetts 02467. E-mail:
| |
Collapse
|
12
|
Delle Bovi RJ, Kim J, Suresh P, London E, Miller WT. Sterol structure dependence of insulin receptor and insulin-like growth factor 1 receptor activation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:819-826. [PMID: 30682326 DOI: 10.1016/j.bbamem.2019.01.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/21/2018] [Accepted: 01/21/2019] [Indexed: 01/02/2023]
Abstract
The plasma membrane is a dynamic environment with a complex composition of lipids, proteins, and cholesterol. Areas enriched in cholesterol and sphingolipids are believed to form lipid rafts, domains of highly ordered lipids. The unique physical properties of these domains have been proposed to influence many cellular processes. Here, we demonstrate that the activation of insulin receptor (IR) and insulin-like growth factor 1 receptor (IGF1R) depends critically on the structures of membrane sterols. IR and IGF1R autophosphorylation in vivo was inhibited by cholesterol depletion, and autophosphorylation was restored by the replacement with exogenous cholesterol. We next screened a variety of sterols for effects on IR activation. The ability of sterols to support IR autophosphorylation was strongly correlated to the propensity of the sterols to form ordered domains. IR autophosphorylation was fully restored by the incorporation of ergosterol, dihydrocholesterol, 7-dehydrocholesterol, lathosterol, desmosterol, and allocholesterol, partially restored by epicholesterol, and not restored by lanosterol, coprostanol, and 4-cholesten-3-one. These data support the hypothesis that the ability to form ordered domains is sufficient for a sterol to support ligand-induced activation of IR and IGF1R in intact mammalian cells.
Collapse
Affiliation(s)
- Richard J Delle Bovi
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794-8661, United States of America
| | - JiHyun Kim
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, United States of America
| | - Pavana Suresh
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, United States of America
| | - Erwin London
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, United States of America.
| | - W Todd Miller
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794-8661, United States of America; Department of Veterans Affairs Medical Center, Northport, NY 11768, United States of America.
| |
Collapse
|
13
|
Key CCC, Liu M, Kurtz CL, Chung S, Boudyguina E, Dinh TA, Bashore A, Phelan PE, Freedman BI, Osborne TF, Zhu X, Ma L, Sethupathy P, Biddinger SB, Parks JS. Hepatocyte ABCA1 Deletion Impairs Liver Insulin Signaling and Lipogenesis. Cell Rep 2018; 19:2116-2129. [PMID: 28591582 DOI: 10.1016/j.celrep.2017.05.032] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 03/07/2017] [Accepted: 05/09/2017] [Indexed: 10/19/2022] Open
Abstract
Plasma membrane (PM) free cholesterol (FC) is emerging as an important modulator of signal transduction. Here, we show that hepatocyte-specific knockout (HSKO) of the cellular FC exporter, ATP-binding cassette transporter A1 (ABCA1), leads to decreased PM FC content and defective trafficking of lysosomal FC to the PM. Compared with controls, chow-fed HSKO mice had reduced hepatic (1) insulin-stimulated Akt phosphorylation, (2) activation of the lipogenic transcription factor Sterol Regulatory Element Binding Protein (SREBP)-1c, and (3) lipogenic gene expression. Consequently, Western-type diet-fed HSKO mice were protected from steatosis. Surprisingly, HSKO mice had intact glucose metabolism; they showed normal gluconeogenic gene suppression in response to re-feeding and normal glucose and insulin tolerance. We conclude that: (1) ABCA1 maintains optimal hepatocyte PM FC, through intracellular FC trafficking, for efficient insulin signaling; and (2) hepatocyte ABCA1 deletion produces a form of selective insulin resistance so that lipogenesis is suppressed but glucose metabolism remains normal.
Collapse
Affiliation(s)
- Chia-Chi C Key
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Mingxia Liu
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - C Lisa Kurtz
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Soonkyu Chung
- Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE 68588, USA
| | - Elena Boudyguina
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Timothy A Dinh
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alexander Bashore
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Peter E Phelan
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA
| | - Barry I Freedman
- Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Timothy F Osborne
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA
| | - Xuewei Zhu
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Lijun Ma
- Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Praveen Sethupathy
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sudha B Biddinger
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02062, USA
| | - John S Parks
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
14
|
Montgomery MK, Brown SHJ, Mitchell TW, Coster ACF, Cooney GJ, Turner N. Association of muscle lipidomic profile with high-fat diet-induced insulin resistance across five mouse strains. Sci Rep 2017; 7:13914. [PMID: 29066734 PMCID: PMC5654831 DOI: 10.1038/s41598-017-14214-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/05/2017] [Indexed: 12/20/2022] Open
Abstract
Different mouse strains exhibit variation in their inherent propensities to develop metabolic disease. We recently showed that C57BL6, 129X1, DBA/2 and FVB/N mice are all susceptible to high-fat diet-induced glucose intolerance, while BALB/c mice are relatively protected, despite changes in many factors linked with insulin resistance. One parameter strongly linked with insulin resistance is ectopic lipid accumulation, especially metabolically active ceramides and diacylglycerols (DAG). This study examined diet-induced changes in the skeletal muscle lipidome across these five mouse strains. High-fat feeding increased total muscle triacylglycerol (TAG) content, with elevations in similar triacylglycerol species observed for all strains. There were also generally consistent changes across strains in the abundance of different phospholipid (PL) classes and the fatty acid profile of phospholipid molecular species, with the exception being a strain-specific difference in phospholipid species containing two polyunsaturated fatty acyl chains in BALB/c mice (i.e. a diet-induced decrease in the other four strains, but no change in BALB/c mice). In contrast to TAG and PL, the high-fat diet had a minor influence on DAG and ceramide species across all strains. These results suggest that widespread alterations in muscle lipids are unlikely a major contributors to the favourable metabolic profile of BALB/c mice and rather there is a relatively conserved high-fat diet response in muscle of most mouse strains.
Collapse
Affiliation(s)
- Magdalene K Montgomery
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
- Diabetes & Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Simon H J Brown
- School of Medicine, University of Wollongong, Wollongong, NSW, Australia
- llawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Todd W Mitchell
- School of Medicine, University of Wollongong, Wollongong, NSW, Australia
- llawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Adelle C F Coster
- School of Mathematics and Statistics, University of New South Wales, Sydney, NSW, Australia
| | - Gregory J Cooney
- Diabetes & Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Nigel Turner
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.
- Diabetes & Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.
| |
Collapse
|
15
|
Zhou X, Shentu P, Xu Y. Spatiotemporal Regulators for Insulin-Stimulated GLUT4 Vesicle Exocytosis. J Diabetes Res 2017; 2017:1683678. [PMID: 28529958 PMCID: PMC5424486 DOI: 10.1155/2017/1683678] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/21/2017] [Accepted: 04/03/2017] [Indexed: 11/30/2022] Open
Abstract
Insulin increases glucose uptake and storage in muscle and adipose cells, which is accomplished through the mobilization of intracellular GLUT4 storage vesicles (GSVs) to the cell surface upon stimulation. Importantly, the dysfunction of insulin-regulated GLUT4 trafficking is strongly linked with peripheral insulin resistance and type 2 diabetes in human. The insulin signaling pathway, key signaling molecules involved, and precise trafficking itinerary of GSVs are largely identified. Understanding the interaction between insulin signaling molecules and key regulatory proteins that are involved in spatiotemporal regulation of GLUT4 vesicle exocytosis is of great importance to explain the pathogenesis of diabetes and may provide new potential therapeutic targets.
Collapse
Affiliation(s)
- Xiaoxu Zhou
- Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Ping Shentu
- Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Yingke Xu
- Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
- *Yingke Xu:
| |
Collapse
|
16
|
Lu JC, Chiang YT, Lin YC, Chang YT, Lu CY, Chen TY, Yeh CS. Disruption of Lipid Raft Function Increases Expression and Secretion of Monocyte Chemoattractant Protein-1 in 3T3-L1 Adipocytes. PLoS One 2016; 11:e0169005. [PMID: 28030645 PMCID: PMC5193455 DOI: 10.1371/journal.pone.0169005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 12/11/2016] [Indexed: 12/13/2022] Open
Abstract
The adipocyte is unique in its capacity to store lipids. In addition to triglycerides, the adipocyte stores a significant amount of cholesterol. Moreover, obese adipocytes are characterized by a redistribution of cholesterol with depleted cholesterol in the plasma membrane, suggesting that cholesterol perturbation may play a role in adipocyte dysfunction. We used methyl-β-cyclodextrin (MβCD), a molecule with high affinity for cholesterol, to rapidly deplete cholesterol level in differentiated 3T3-L1 adipocytes. We tested whether this perturbation altered adipocyte secretion of monocyte chemoattractant protein-1 (MCP-1), a chemokine that is elevated in obesity and is linked to obesity-associated chronic diseases. Depletion of cholesterol by MβCD increased MCP-1 secretion as well as the mRNA and protein levels, suggesting perturbation at biosynthesis and secretion. Pharmacological inhibition revealed that NF-κB, but not MEK, p38 and JNK, was involved in MβCD-stimulated MCP-1 biosynthesis and secretion in adipocytes. Finally, another cholesterol-binding drug, filipin, also induced MCP-1 secretion without altering membrane cholesterol level. Interestingly, both MβCD and filipin disturbed the integrity of lipid rafts, the membrane microdomains enriched in cholesterol. Thus, the depletion of membrane cholesterol in obese adipocytes may result in dysfunction of lipid rafts, leading to the elevation of proinflammatory signaling and MCP-1 secretion in adipocytes.
Collapse
Affiliation(s)
- Juu-Chin Lu
- Department of Physiology and Pharmacology, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
- * E-mail:
| | - Yu-Ting Chiang
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Chun Lin
- Department of Physiology and Pharmacology, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Tzu Chang
- Department of Physiology and Pharmacology, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Yun Lu
- Department of Physiology and Pharmacology, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Tzu-Yu Chen
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Shan Yeh
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
17
|
Montgomery MK, Brown SHJ, Lim XY, Fiveash CE, Osborne B, Bentley NL, Braude JP, Mitchell TW, Coster ACF, Don AS, Cooney GJ, Schmitz-Peiffer C, Turner N. Regulation of glucose homeostasis and insulin action by ceramide acyl-chain length: A beneficial role for very long-chain sphingolipid species. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1828-1839. [PMID: 27591968 DOI: 10.1016/j.bbalip.2016.08.016] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 08/18/2016] [Accepted: 08/29/2016] [Indexed: 12/13/2022]
Abstract
In a recent study, we showed that in response to high fat feeding C57BL/6, 129X1, DBA/2 and FVB/N mice all developed glucose intolerance, while BALB/c mice displayed minimal deterioration in glucose tolerance and insulin action. Lipidomic analysis of livers across these five strains has revealed marked strain-specific differences in ceramide (Cer) and sphingomyelin (SM) species with high-fat feeding; with increases in C16-C22 (long-chain) and reductions in C>22 (very long-chain) Cer and SM species observed in the four strains that developed HFD-induced glucose intolerance. Intriguingly, the opposite pattern was observed in sphingolipid species in BALB/c mice. These strain-specific changes in sphingolipid acylation closely correlated with ceramide synthase 2 (CerS2) protein content and activity, with reduced CerS2 levels/activity observed in glucose intolerant strains and increased content in BALB/c mice. Overexpression of CerS2 in primary mouse hepatocytes induced a specific elevation in very long-chain Cer, but despite the overall increase in ceramide abundance, there was a substantial improvement in insulin signal transduction, as well as decreased ER stress and gluconeogenic markers. Overall our findings suggest that very long-chain sphingolipid species exhibit a protective role against the development of glucose intolerance and hepatic insulin resistance.
Collapse
Affiliation(s)
- Magdalene K Montgomery
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | - Simon H J Brown
- School of Medicine, University of Wollongong, Wollongong, NSW, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Xin Y Lim
- Prince of Wales Clinical School, UNSW Australia, Sydney, NSW, Australia
| | - Corrine E Fiveash
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | - Brenna Osborne
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | - Nicholas L Bentley
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | - Jeremy P Braude
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | - Todd W Mitchell
- School of Medicine, University of Wollongong, Wollongong, NSW, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Adelle C F Coster
- School of Mathematics and Statistics, UNSW Australia, Sydney, NSW, Australia
| | - Anthony S Don
- Prince of Wales Clinical School, UNSW Australia, Sydney, NSW, Australia
| | - Gregory J Cooney
- Diabetes & Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St Vincent's Clinical School, UNSW Australia, Sydney, NSW, Australia
| | - Carsten Schmitz-Peiffer
- Diabetes & Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St Vincent's Clinical School, UNSW Australia, Sydney, NSW, Australia
| | - Nigel Turner
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia.
| |
Collapse
|
18
|
Nagaraj V, Kazim AS, Helgeson J, Lewold C, Barik S, Buda P, Reinbothe TM, Wennmalm S, Zhang E, Renström E. Elevated Basal Insulin Secretion in Type 2 Diabetes Caused by Reduced Plasma Membrane Cholesterol. Mol Endocrinol 2016; 30:1059-1069. [PMID: 27533789 PMCID: PMC5045496 DOI: 10.1210/me.2016-1023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Elevated basal insulin secretion under fasting conditions together with insufficient stimulated insulin release is an important hallmark of type 2 diabetes, but the mechanisms controlling basal insulin secretion remain unclear. Membrane rafts exist in pancreatic islet cells and spatially organize membrane ion channels and proteins controlling exocytosis, which may contribute to the regulation of insulin secretion. Membrane rafts (cholesterol and sphingolipid containing microdomains) were dramatically reduced in human type 2 diabetic and diabetic Goto-Kakizaki (GK) rat islets when compared with healthy islets. Oxidation of membrane cholesterol markedly reduced microdomain staining intensity in healthy human islets, but was without effect in type 2 diabetic islets. Intriguingly, oxidation of cholesterol affected glucose-stimulated insulin secretion only modestly, whereas basal insulin release was elevated. This was accompanied by increased intracellular Ca2+ spike frequency and Ca2+ influx and explained by enhanced single Ca2+ channel activity. These results suggest that the reduced presence of membrane rafts could contribute to the elevated basal insulin secretion seen in type 2 diabetes.
Collapse
Affiliation(s)
- Vini Nagaraj
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Abdulla S Kazim
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Johan Helgeson
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Clemens Lewold
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Satadal Barik
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Pawel Buda
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Thomas M Reinbothe
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Stefan Wennmalm
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Enming Zhang
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Erik Renström
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| |
Collapse
|
19
|
Abstract
Endocytosis can be separated into the categories of phagocytosis and pinocytosis. Phagocytosis can be distinguished from pinocytosis primarily by the size of particle ingested and by its dependence on actin polymerization as a key step in particle ingestion. Several specific forms of pinocytosis have been identified that can be distinguished based on their dependence on clathrin or caveolin. Both clathrin and caveolin-dependent pinocytosis appear to require the participation of dynamin to internalize the plasma membrane. Other, less well-characterized forms of pinocytosis have also been described. Although endocytosis has long been known to affect receptor density, recent studies have demonstrated that endocytosis through clathrin- and caveolin-dependent processes plays a key role in receptor-mediated signal transduction. In some cases, blockade of these processes attenuates, or even prevents, signal transduction from taking place. This information, coupled with a better understanding of endocytosis mechanisms, will help advance the field of cell biology as well as present new targets for drug development and disease treatment.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Medicine, Room 12, Ruppert Center, 3120 Glendale Avenue, Toledo, OH 43614, USA.
| | | |
Collapse
|
20
|
Escribá PV, Busquets X, Inokuchi JI, Balogh G, Török Z, Horváth I, Harwood JL, Vígh L. Membrane lipid therapy: Modulation of the cell membrane composition and structure as a molecular base for drug discovery and new disease treatment. Prog Lipid Res 2015; 59:38-53. [PMID: 25969421 DOI: 10.1016/j.plipres.2015.04.003] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 04/10/2015] [Accepted: 04/29/2015] [Indexed: 01/17/2023]
Abstract
Nowadays we understand cell membranes not as a simple double lipid layer but as a collection of complex and dynamic protein-lipid structures and microdomains that serve as functional platforms for interacting signaling lipids and proteins. Membrane lipids and lipid structures participate directly as messengers or regulators of signal transduction. In addition, protein-lipid interactions participate in the localization of signaling protein partners to specific membrane microdomains. Thus, lipid alterations change cell signaling that are associated with a variety of diseases including cancer, obesity, neurodegenerative disorders, cardiovascular pathologies, etc. This article reviews the newly emerging field of membrane lipid therapy which involves the pharmacological regulation of membrane lipid composition and structure for the treatment of diseases. Membrane lipid therapy proposes the use of new molecules specifically designed to modify membrane lipid structures and microdomains as pharmaceutical disease-modifying agents by reversing the malfunction or altering the expression of disease-specific protein or lipid signal cascades. Here, we provide an in-depth analysis of this emerging field, especially its molecular bases and its relevance to the development of innovative therapeutic approaches.
Collapse
Affiliation(s)
- Pablo V Escribá
- Department of Biology, University of the Balearic Islands, E-07122 Palma de Mallorca, Spain
| | - Xavier Busquets
- Department of Biology, University of the Balearic Islands, E-07122 Palma de Mallorca, Spain
| | - Jin-ichi Inokuchi
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai, Japan
| | - Gábor Balogh
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary
| | - Zsolt Török
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary
| | - Ibolya Horváth
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary
| | - John L Harwood
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, Wales, UK.
| | - László Vígh
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, Szeged, Hungary.
| |
Collapse
|
21
|
Rostoker R, Abelson S, Bitton-Worms K, Genkin I, Ben-Shmuel S, Dakwar M, Orr ZS, Caspi A, Tzukerman M, LeRoith D. Highly specific role of the insulin receptor in breast cancer progression. Endocr Relat Cancer 2015; 22:145-57. [PMID: 25694511 PMCID: PMC4362669 DOI: 10.1530/erc-14-0490] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Accumulating evidence from clinical trials indicates that specific targeting of the IGF1 receptor (IGF1R) is not efficient as an anti-breast cancer treatment. One possible reason is that the mitogenic signals from the insulin receptor (IR) can be processed independently or as compensation to inhibition of the IGF1R. In this study, we highlight the role of the IR in mediating breast tumor progression in both WT mice and a hyperinsulinemic MKR mouse model by induction of Ir (Insr) or Igf1r knockdown (KD) in the mammary carcinoma Mvt-1 cell line. By using the specific IR antagonist-S961, we demonstrated that Igf1r-KD induces elevated responses by the IR to IGF1. On the other hand, Ir-KD cells generated significantly smaller tumors in the mammary fat pads of both WT and MKR mice, as opposed to control cells, whereas the Igf1r-KD cells did not. The tumorigenic effects of insulin on the Mvt-1 cells were also demonstrated using microarray analysis, which indicates alteration of genes and signaling pathways involved in proliferation, the cell cycle, and apoptosis following insulin stimulation. In addition, the correlation between IR and the potential prognostic marker for aggressive breast cancer, CD24, was examined in the Ir-KD cells. Fluorescence-activated cell sorting (FACS) analysis revealed more than 60% reduction in CD24 expression in the Ir-KD cells when compared with the control cells. Our results also indicate that CD24-expressing cells can restore, at least in part, the tumorigenic capacity of Ir-KD cells. Taken together, our results highlight the mitogenic role of the IR in mammary tumor progression with a direct link to CD24 expression.
Collapse
Affiliation(s)
- Ran Rostoker
- Clinical Research Institute at Rambam (CRIR) and the Faculty of MedicineTechnion, Diabetes and Metabolism Clinical Research Center of Excellence, Haifa, IsraelThe Laboratory of Molecular MedicineRambam Health Care Campus and Rappaport Faculty of Medicine and Research Institute, Technion, Haifa 31096, IsraelDivision of EndocrinologyDiabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Sagi Abelson
- Clinical Research Institute at Rambam (CRIR) and the Faculty of MedicineTechnion, Diabetes and Metabolism Clinical Research Center of Excellence, Haifa, IsraelThe Laboratory of Molecular MedicineRambam Health Care Campus and Rappaport Faculty of Medicine and Research Institute, Technion, Haifa 31096, IsraelDivision of EndocrinologyDiabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Keren Bitton-Worms
- Clinical Research Institute at Rambam (CRIR) and the Faculty of MedicineTechnion, Diabetes and Metabolism Clinical Research Center of Excellence, Haifa, IsraelThe Laboratory of Molecular MedicineRambam Health Care Campus and Rappaport Faculty of Medicine and Research Institute, Technion, Haifa 31096, IsraelDivision of EndocrinologyDiabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Inna Genkin
- Clinical Research Institute at Rambam (CRIR) and the Faculty of MedicineTechnion, Diabetes and Metabolism Clinical Research Center of Excellence, Haifa, IsraelThe Laboratory of Molecular MedicineRambam Health Care Campus and Rappaport Faculty of Medicine and Research Institute, Technion, Haifa 31096, IsraelDivision of EndocrinologyDiabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Sarit Ben-Shmuel
- Clinical Research Institute at Rambam (CRIR) and the Faculty of MedicineTechnion, Diabetes and Metabolism Clinical Research Center of Excellence, Haifa, IsraelThe Laboratory of Molecular MedicineRambam Health Care Campus and Rappaport Faculty of Medicine and Research Institute, Technion, Haifa 31096, IsraelDivision of EndocrinologyDiabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Maria Dakwar
- Clinical Research Institute at Rambam (CRIR) and the Faculty of MedicineTechnion, Diabetes and Metabolism Clinical Research Center of Excellence, Haifa, IsraelThe Laboratory of Molecular MedicineRambam Health Care Campus and Rappaport Faculty of Medicine and Research Institute, Technion, Haifa 31096, IsraelDivision of EndocrinologyDiabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Zila Shen Orr
- Clinical Research Institute at Rambam (CRIR) and the Faculty of MedicineTechnion, Diabetes and Metabolism Clinical Research Center of Excellence, Haifa, IsraelThe Laboratory of Molecular MedicineRambam Health Care Campus and Rappaport Faculty of Medicine and Research Institute, Technion, Haifa 31096, IsraelDivision of EndocrinologyDiabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Avishay Caspi
- Clinical Research Institute at Rambam (CRIR) and the Faculty of MedicineTechnion, Diabetes and Metabolism Clinical Research Center of Excellence, Haifa, IsraelThe Laboratory of Molecular MedicineRambam Health Care Campus and Rappaport Faculty of Medicine and Research Institute, Technion, Haifa 31096, IsraelDivision of EndocrinologyDiabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Maty Tzukerman
- Clinical Research Institute at Rambam (CRIR) and the Faculty of MedicineTechnion, Diabetes and Metabolism Clinical Research Center of Excellence, Haifa, IsraelThe Laboratory of Molecular MedicineRambam Health Care Campus and Rappaport Faculty of Medicine and Research Institute, Technion, Haifa 31096, IsraelDivision of EndocrinologyDiabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Derek LeRoith
- Clinical Research Institute at Rambam (CRIR) and the Faculty of MedicineTechnion, Diabetes and Metabolism Clinical Research Center of Excellence, Haifa, IsraelThe Laboratory of Molecular MedicineRambam Health Care Campus and Rappaport Faculty of Medicine and Research Institute, Technion, Haifa 31096, IsraelDivision of EndocrinologyDiabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York City, New York, USA Clinical Research Institute at Rambam (CRIR) and the Faculty of MedicineTechnion, Diabetes and Metabolism Clinical Research Center of Excellence, Haifa, IsraelThe Laboratory of Molecular MedicineRambam Health Care Campus and Rappaport Faculty of Medicine and Research Institute, Technion, Haifa 31096, IsraelDivision of EndocrinologyDiabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| |
Collapse
|
22
|
Peng Q, Jia SH, Parodo J, Ai Y, Marshall JC. Pre-B cell colony enhancing factor induces Nampt-dependent translocation of the insulin receptor out of lipid microdomains in A549 lung epithelial cells. Am J Physiol Endocrinol Metab 2015; 308:E324-33. [PMID: 25516545 DOI: 10.1152/ajpendo.00006.2014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pre-B cell colony-enhancing factor (PBEF) is a highly conserved pleiotropic protein reported to be an alternate ligand for the insulin receptor (IR). We sought to clarify the relationship between PBEF and insulin signaling by evaluating the effects of PBEF on the localization of the IRβ chain to lipid rafts in A549 epithelial cells. We isolated lipid rafts from A549 cells and detected the IR by immunoprecipitation from raft fractions or whole cell lysates. Cells were treated with rPBEF, its enzymatic product nicotinamide adenine dinucleotide (NAD), or the Nampt inhibitor daporinad to study the effect of PBEF on IRβ movement. We used coimmunoprecipitation studies in cells transfected with PBEF and IRβ constructs to detect interactions between PBEF, the IRβ, and caveolin-1 (Cav-1). PBEF was present in both lipid raft and nonraft fractions, whereas the IR was found only in lipid raft fractions of resting A549 cells. The IR-, PBEF-, and Cav-1-coimmunoprecipitated rPBEF treatment resulted in the movement of IRβ- and tyrosine-phosphorylated Cav-1 from lipid rafts to nonrafts, an effect that could be blocked by daporinad, suggesting that this effect was facilitated by the Nampt activity of PBEF. The addition of PBEF to insulin-treated cells resulted in reduced Akt phosphorylation of both Ser⁴⁷³ and Thr³⁰⁸. We conclude that PBEF can inhibit insulin signaling through the IR by Nampt-dependent promotion of IR translocation into the nonraft domains of A549 epithelial cells. PBEF-induced alterations in the spatial geometry of the IR provide a mechanistic explanation for insulin resistance in inflammatory states associated with upregulation of PBEF.
Collapse
Affiliation(s)
- Qianyi Peng
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and the Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada; and Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Song Hui Jia
- Department of Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and the Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada; and
| | - Jean Parodo
- Department of Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and the Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada; and
| | - Yuhang Ai
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - John C Marshall
- Department of Surgery, Department of Critical Care Medicine, and Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and the Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada; and
| |
Collapse
|
23
|
Onopchenko OV. The effect of N-stearoylethanolamine on the activity of antioxidant enzymes, content of lipid peroxidation products and nitric oxide in the blood plasma and liver of rats with induced insulin-resistance. UKRAINIAN BIOCHEMICAL JOURNAL 2013. [DOI: 10.15407/ubj85.05.088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
24
|
Elvira B, Honisch S, Almilaji A, Pakladok T, Liu G, Shumilina E, Alesutan I, Yang W, Munoz C, Lang F. Up-regulation of Na(+)-coupled glucose transporter SGLT1 by caveolin-1. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:2394-8. [PMID: 23774524 DOI: 10.1016/j.bbamem.2013.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 05/29/2013] [Accepted: 06/06/2013] [Indexed: 01/09/2023]
Abstract
The Na(+)-coupled glucose transporter SGLT1 (SLC5A1) accomplishes concentrative cellular glucose uptake even at low extracellular glucose concentrations. The carrier is expressed in renal proximal tubules, small intestine and a variety of nonpolarized cells including several tumor cells. The present study explored whether SGLT1 activity is regulated by caveolin-1, which is known to regulate the insertion of several ion channels and carriers in the cell membrane. To this end, SGLT1 was expressed in Xenopus oocytes with or without additional expression of caveolin-1 and electrogenic glucose transport determined by dual electrode voltage clamp experiments. In SGLT1-expressing oocytes, but not in oocytes injected with water or caveolin-1 alone, the addition of glucose to the extracellular bath generated an inward current (Ig), which was increased following coexpression of caveolin-1. Kinetic analysis revealed that caveolin-1 increased maximal Ig without significantly modifying the glucose concentration required to trigger half maximal Ig (KM). According to chemiluminescence and confocal microscopy, caveolin-1 increased SGLT1 protein abundance in the cell membrane. Inhibition of SGLT1 insertion by brefeldin A (5μM) resulted in a decline of Ig, which was similar in the absence and presence of caveolin-1. In conclusion, caveolin-1 up-regulates SGLT1 activity by increasing carrier protein abundance in the cell membrane, an effect presumably due to stimulation of carrier protein insertion into the cell membrane.
Collapse
Affiliation(s)
- Bernat Elvira
- Department of Physiology, University of Tübingen, Gmelinstr. 5, D-72076 Tübingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Perez-Cervera Y, Dehennaut V, Aquino Gil M, Guedri K, Solórzano Mata CJ, Olivier-Van Stichelen S, Michalski JC, Foulquier F, Lefebvre T. Insulin signaling controls the expression of O-GlcNAc transferase and its interaction with lipid microdomains. FASEB J 2013; 27:3478-86. [PMID: 23689613 DOI: 10.1096/fj.12-217984] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Lipid microdomains (rafts) are cholesterol-enriched dynamic ordered lipid domains belonging to cell membranes involved in diverse cellular functions, including signal transduction, membrane trafficking, and infection. Many studies have reported relationships between insulin signaling and lipid rafts. Likewise, links between insulin signaling and O-GlcNAcylation have also been described. However, the potential connection between O-GlcNAc and raft dynamics remains unexplored. Here we show that O-GlcNAc and the enzyme that creates this modification, O-GlcNAc transferase (OGT), are localized in rafts. On insulin stimulation, we observe time-dependent increases in OGT expression and localization within rafts. We show that these processes depend on activation of the phosphatidylinositol 3-kinase (PI3K) pathway. Inhibition of OGT does not significantly affect cholesterol synthesis and raft building but decreases insulin receptor expression and PI3K and mitogen-activated protein kinase pathway activation. Taken together, these findings indicate that O-GlcNAcylation, lipid rafts, and signaling pathways are spatiotemporally coordinated to enable fundamental cellular functions.
Collapse
Affiliation(s)
- Yobana Perez-Cervera
- Centre National de la Recherche Scientifique (CNRS)–Unité Mixte de Recherche (UMR) 8576, Institut Fédératif de Recherche (IFR) 147, University of Lille 1, Villeneuve d'Ascq, France
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Pedersen DJ, Diakanastasis B, Stöckli J, Schmitz-Peiffer C. Protein kinase Cε modulates insulin receptor localization and trafficking in mouse embryonic fibroblasts. PLoS One 2013; 8:e58046. [PMID: 23469261 PMCID: PMC3585804 DOI: 10.1371/journal.pone.0058046] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 01/30/2013] [Indexed: 12/03/2022] Open
Abstract
We have previously shown that deletion of protein kinase C epsilon (PKCε) in mice results in protection against glucose intolerance caused by a high fat diet. This was in part due to reduced insulin uptake by hepatocytes and insulin clearance, which enhanced insulin availability. Here we employed mouse embryonic fibroblasts (MEFs) derived from wildtype (WT) and PKCε-deficient (PKCε−/−) mice to examine this mechanistically. PKCε−/− MEFs exhibited reduced insulin uptake which was associated with decreased insulin receptor phosphorylation, while downstream signalling through IRS-1 and Akt was unaffected. Cellular fractionation demonstrated that PKCε deletion changed the localization of the insulin receptor, a greater proportion of which co-fractionated with flotillin-1, a marker of membrane microdomains. Insulin stimulation resulted in redistribution of the receptor in WT cells, while this was markedly reduced in PKCε−/− cells. These alterations in insulin receptor trafficking were associated with reduced expression of CEACAM1, a receptor substrate previously shown to modulate insulin clearance. Virally-mediated reconstitution of PKCε in MEFs increased CEACAM1 expression and partly restored the sensitivity of the receptor to insulin-stimulated redistribution. These data indicate that PKCε can affect insulin uptake in MEFs through promotion of receptor-mediated endocytosis, and that this may be mediated by regulation of CEACAM1 expression.
Collapse
Affiliation(s)
- David J. Pedersen
- Diabetes and Obesity Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Barbara Diakanastasis
- Diabetes and Obesity Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Jacqueline Stöckli
- Diabetes and Obesity Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Carsten Schmitz-Peiffer
- Diabetes and Obesity Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
27
|
Tekpli X, Holme JA, Sergent O, Lagadic-Gossmann D. Role for membrane remodeling in cell death: Implication for health and disease. Toxicology 2013; 304:141-57. [DOI: 10.1016/j.tox.2012.12.014] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 11/29/2012] [Accepted: 12/20/2012] [Indexed: 12/31/2022]
|
28
|
Park JW, Park WJ, Kuperman Y, Boura-Halfon S, Pewzner-Jung Y, Futerman AH. Ablation of very long acyl chain sphingolipids causes hepatic insulin resistance in mice due to altered detergent-resistant membranes. Hepatology 2013; 57:525-32. [PMID: 22911490 DOI: 10.1002/hep.26015] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 08/08/2012] [Indexed: 12/11/2022]
Abstract
UNLABELLED Sphingolipids are important structural components of cell membranes and act as critical regulators of cell function by modulating intracellular signaling pathways. Specific sphingolipids, such as ceramide, glucosylceramide, and ganglioside GM3, have been implicated in various aspects of insulin resistance, because they have been shown to modify several steps in the insulin signaling pathway, such as phosphorylation of either protein kinase B (Akt) or of the insulin receptor. We now explore the role of the ceramide acyl chain length in insulin signaling by using a ceramide synthase 2 (CerS2) null mouse, which is unable to synthesize very long acyl chain (C22-C24) ceramides. CerS2 null mice exhibited glucose intolerance despite normal insulin secretion from the pancreas. Both insulin receptor and Akt phosphorylation were abrogated in liver, but not in adipose tissue or in skeletal muscle. The lack of insulin receptor phosphorylation in liver correlated with its inability to translocate into detergent-resistant membranes (DRMs). Moreover, DRMs in CerS2 null mice displayed properties significantly different from those in wild-type mice, suggesting that the altered sphingolipid acyl chain length directly affects insulin receptor translocation and subsequent signaling. CONCLUSION We conclude that the sphingolipid acyl chain composition of liver regulates insulin signaling by modifying insulin receptor translocation into membrane microdomains.
Collapse
Affiliation(s)
- Joo-Won Park
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | | | |
Collapse
|
29
|
Wu Y, Chen X, Wang S, Jiang M, Zheng B, Zhou Q, Bi Y, Zhou Z, Huang X, Sha J. Flotillin-2 is an acrosome-related protein involved in mouse spermiogenesis. J Biomed Res 2012; 26:278-87. [PMID: 23554761 PMCID: PMC3596745 DOI: 10.7555/jbr.26.20120030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 04/27/2012] [Accepted: 05/03/2012] [Indexed: 11/28/2022] Open
Abstract
Spermatogenesis is a complex process of terminal differentiation by which mature sperms are generated, and it can be divided into three phases: mitosis, meiosis and spermiogenesis. In a previous study, we established a series of proteomic profiles for spermatogenesis to understand the regulation of male fertility and infertility. Here, we further investigated the localization and the role of flotillin-2 in spermiogenesis. Flotillin-2 expression was investigated in the testis of male CD1 mice at various developmental stages of spermatogenesis by using Western blotting, immunohistochemistry and immunofluorescence. Flotillin-2 was knocked down in vivo in three-week-old male mice using intratesticular injection of small inhibitory RNA (siRNA), and sperm abnormalities were assessed three weeks later. Flotillin-2 was expressed at high levels in male germ cells during spermatogenesis. Flotillin-2 immunoreactivity was observed in pachytene spermatocytes as a strong dot-shaped signal and in round spermatids as a sickle-shaped distribution ahead of the acrosome. Immunofluorescence confirmed flotillin-2 was localized in front of the acrosome in round spermatids, indicating that flotillin-2 was localized to the Golgi apparatus. Knockdown of flotillin-2in vivo led to a significant increase in head sperm abnormalities isolated from the cauda epididymis, compared with control siRNA-injected testes. This study indicates that flotillin-2 is a novel Golgi-related protein involved in sperm acrosome biogenesis.
Collapse
Affiliation(s)
- Yibo Wu
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Tan Z, Zhou LJ, Mu PW, Liu SP, Chen SJ, Fu XD, Wang TH. Caveolin-3 is involved in the protection of resveratrol against high-fat-diet-induced insulin resistance by promoting GLUT4 translocation to the plasma membrane in skeletal muscle of ovariectomized rats. J Nutr Biochem 2012; 23:1716-24. [PMID: 22569348 DOI: 10.1016/j.jnutbio.2011.12.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 11/28/2011] [Accepted: 12/13/2011] [Indexed: 01/26/2023]
Abstract
Insulin resistance is recognized as a common metabolic factor which predicts the future development of both type 2 diabetes and atherosclerotic disease. Resveratrol (RSV), an agonist of estrogen receptor (ER), is known to affect insulin sensitivity, but the mechanism is unclear. Evidence suggests that caveolin-3 (CAV-3), a member of the caveolin family, is involved in insulin-stimulated glucose uptake. Our recent work indicated that estrogen via ER improves glucose uptake by up-regulation of CAV-3 expression. Here, we investigated the role of CAV-3 in the effect of RSV on insulin resistance in skeletal muscle both in vivo and in vitro. The results demonstrated that RSV ameliorated high-fat-diet (HFD)-induced glucose intolerance and insulin resistance in ovariectomized rats. RSV elevated insulin-stimulated glucose uptake in isolated soleus muscle in vivo and in C2C12 myotubes in vitro by enhancing GLUT4 translocation to the plasma membrane rather than increasing GLUT4 protein expression. Through ERα-mediated transcription, RSV increased CAV-3 protein expression, which contributed to GLUT4 translocation. Moreover, after knockdown of CAV-3 gene, the effects of RSV on glucose uptake and the translocation of GLUT4 to the plasma membrane, as well as the association of CAV-3 and GLUT4 in the membrane, were significantly attenuated. Our findings demonstrated that RSV via ERα elevated CAV-3 expression and then enhanced GLUT4 translocation to the plasma membrane to promote glucose uptake in skeletal muscle, exerting its protective effects against HFD-induced insulin resistance. It suggests that this pathway could represent an effective therapeutic target to fight against insulin resistance syndrome induced by HFD.
Collapse
Affiliation(s)
- Zhi Tan
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, PR China.
| | | | | | | | | | | | | |
Collapse
|
31
|
Lv Z, Zhang X, Liu L, Chen J, Nie Z, Sheng Q, Zhang W, Jiang C, Yu W, Wang D, Wu X, Zhang S, Li J, Zhang Y. Characterization of a gene encoding prohibitin in silkworm, Bombyx mori. Gene 2012; 502:118-24. [PMID: 22450364 DOI: 10.1016/j.gene.2012.03.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Revised: 02/23/2012] [Accepted: 03/04/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND Prohibitin (PHB) is an evolutionarily conserved multifunctional protein with ubiquitous expression. However, its molecular roles are largely unknown. METHODS To better understand the function of prohibitin protein in silkworm (BmPHB), its coding sequence was isolated from a cDNA library of silkworm pupae. An His-tagged BmPHB fusion protein was expressed in Escherichia coli Rosetta (DE3) and purified with affinity and reversed-phase chromatography. Purified rBmPHB was used to generate anti-BmPHB polyclonal antibody. The subcellular localization of BmPHB was analysed by immunohistochemistry. RESULTS BmPHB gene has an ORF of 825 bp, encoding a predicted peptide with 274 amino acid residues. Immunostaining indicate that prohibitin is expressed in nucleus and predominately in cytoplasm. Western blot analyses indicated that, in the fifth instar larva, BmPHB was expressed descendingly in gonad, malpighian tubule, trachea, fatty body, intestine, and head. However, no expression was detected in larva's silk gland and epidermis. In addition, BmPHB was expressed in the nascent egg, larva and pupa, but not in the moth. CONCLUSIONS The expression of BmPHB gene presents differential characteristic in different stage and tissues. It may play important roles in the development of silkworm. GENERAL SIGNIFICANCE Studies on prohibitin have been still restricted to a few specific insects and insect cell lines such as Drosophila, Acyrthosiphon pisum and mosquito cell lines, not yet in silkworm. This is a first characterization of prohibitin in silkworm, B. mori.
Collapse
Affiliation(s)
- Zhengbing Lv
- Institute of Biochemistry, College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Shen SH, Wertz DL, Klinman JP. Implication for functions of the ectopic adipocyte copper amine oxidase (AOC3) from purified enzyme and cell-based kinetic studies. PLoS One 2012; 7:e29270. [PMID: 22238597 PMCID: PMC3251558 DOI: 10.1371/journal.pone.0029270] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 11/23/2011] [Indexed: 12/23/2022] Open
Abstract
AOC3 is highly expressed in adipocytes and smooth muscle cells, but its function in these cells is currently unknown. The in vivo substrate(s) of AOC3 is/are also unknown, but could provide an invaluable clue to the enzyme's function. Expression of untagged, soluble human AOC3 in insect cells provides a relatively simple means of obtaining pure enzyme. Characterization of enzyme indicates a 6% titer for the active site 2,4,5-trihydroxyphenylalanine quinone (TPQ) cofactor and corrected k(cat) values as high as 7 s(-1). Substrate kinetic profiling shows that the enzyme accepts a variety of primary amines with different chemical features, including nonphysiological branched-chain and aliphatic amines, with measured k(cat)/K(m) values between 10(2) and 10(4) M(-1) s(-1). K(m)(O(2)) approximates the partial pressure of oxygen found in the interstitial space. Comparison of the properties of purified murine to human enzyme indicates k(cat)/K(m) values that are within 3 to 4-fold, with the exception of methylamine and aminoacetone that are ca. 10-fold more active with human AOC3. With drug development efforts investigating AOC3 as an anti-inflammatory target, these studies suggest that caution is called for when screening the efficacy of inhibitors designed against human enzymes in non-transgenic mouse models. Differentiated murine 3T3-L1 adipocytes show a uniform distribution of AOC3 on the cell surface and whole cell K(m) values that are reasonably close to values measured using purified enzymes. The latter studies support a relevance of the kinetic parameters measured with isolated AOC3 variants to adipocyte function. From our studies, a number of possible substrates with relatively high k(cat)/K(m) have been discovered, including dopamine and cysteamine, which may implicate a role for adipocyte AOC3 in insulin-signaling and fatty acid metabolism, respectively. Finally, the demonstrated AOC3 turnover of primary amines that are non-native to human tissue suggests possible roles for the adipocyte enzyme in subcutaneous bacterial infiltration and obesity.
Collapse
Affiliation(s)
- Sam H. Shen
- Department of Chemistry, University of California, Berkeley, California, United States of America
| | - Diana L. Wertz
- Department of Chemistry, University of California, Berkeley, California, United States of America
| | - Judith P. Klinman
- Department of Chemistry, University of California, Berkeley, California, United States of America
- Department of Molecular and Cell Biology and the California Institute for Quantitative Biosciences, University of California, Berkeley, California, United States of America
| |
Collapse
|
33
|
Mascitelli L, Goldstein MR. Long-Standing Statin Therapy and the Risk of New-Onset Diabetes in the Elderly. Drugs Aging 2012; 29:9-13. [DOI: 10.2165/11598530-000000000-00000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
34
|
|
35
|
Inokuchi JI. Physiopathological function of hematoside (GM3 ganglioside). PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2011; 87:179-98. [PMID: 21558756 PMCID: PMC3149380 DOI: 10.2183/pjab.87.179] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Since I was involved in the molecular cloning of GM3 synthase (SAT-I), which is the primary enzyme for the biosynthesis of gangliosides in 1998, my research group has been concentrating on our efforts to explore the physiological and pathological implications of gangliosides especially for GM3. During the course of study, we demonstrated the molecular pathogenesis of type 2 diabetes and insulin resistance focusing on the interaction between insulin receptor and gangliosides in membrane microdomains and propose a new concept: Life style-related diseases, such as type 2 diabetes, are a membrane microdomain disorder caused by aberrant expression of gangliosides. We also encountered an another interesting aspect indicating the indispensable role of gangliosides in auditory system. After careful behavioral examinations of SAT-I knockout mice, their hearing ability was seriously impaired with selective degeneration of the stereocilia of hair cells in the organ of Corti. This is the first observation demonstrating a direct link between gangliosides and hearing functions.
Collapse
Affiliation(s)
- Jin-ichi Inokuchi
- Division of Glycopathology, Institute of Molecular Biomembranes and Glycobiology, Tohoku Pharmaceutical University, Miyagi, Japan.
| |
Collapse
|
36
|
Inhibition of ganglioside biosynthesis as a novel therapeutic approach in insulin resistance. Handb Exp Pharmacol 2011:165-78. [PMID: 21484572 DOI: 10.1007/978-3-642-17214-4_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
A new concept "Life style-related diseases, such as type 2 diabetes, are a membrane microdomain disorder caused by aberrant expression of gangliosides" has arisen. By examining this working hypothesis, we demonstrate the molecular pathogenesis of type 2 diabetes and insulin resistance focusing on the interaction between insulin receptor and gangliosides in microdomains microdomains and propose the new therapeutic strategy "membrane microdomain ortho-signaling therapy".
Collapse
|
37
|
Fuller M. Sphingolipids: the nexus between Gaucher disease and insulin resistance. Lipids Health Dis 2010; 9:113. [PMID: 20937139 PMCID: PMC2964722 DOI: 10.1186/1476-511x-9-113] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Accepted: 10/11/2010] [Indexed: 12/12/2022] Open
Abstract
Sphingolipids constitute a diverse array of lipids in which fatty acids are linked through amide bonds to a long-chain base, and, structurally, they form the building blocks of eukaryotic membranes. Ceramide is the simplest and serves as a precursor for the synthesis of the three main types of complex sphingolipids; sphingomyelins, glycosphingolipids and gangliosides. Sphingolipids are no longer considered mere structural spectators, but bioactive molecules with functions beyond providing a mechanically stable and chemically resistant barrier to a diverse array of cellular processes. Although sphingolipids form a somewhat minor component of the total cellular lipid pool, their accumulation in certain cells forms the basis of many diseases. Human diseases caused by alterations in the metabolism of sphingolipids are conventionally inborn errors of degradation, the most common being Gaucher disease, in which the catabolism of glucosylceramide is defective and accumulates. Insulin resistance has been reported in patients with Gaucher disease and this article presents evidence that this is due to perturbations in the metabolism of sphingolipids. Ceramide and the more complex sphingolipids, the gangliosides, are constituents of specialised membrane microdomains termed lipid rafts. Lipid rafts play a role in facilitating and regulating lipid and protein interactions in cells, and their unique lipid composition enables them to carry out this role. The lipid composition of rafts is altered in cell models of Gaucher disease which may be responsible for impaired lipid and protein sorting observed in this disorder, and consequently pathology. Lipid rafts are also necessary for correct insulin signalling, and a perturbed lipid raft composition may impair insulin signalling. Unravelling common nodes of interaction between insulin resistance and Gaucher disease may lead to a better understanding of the biochemical mechanisms behind pathology.
Collapse
Affiliation(s)
- Maria Fuller
- Lysosomal Diseases Research Unit, Genetics and Molecular Pathology, SA Pathology, Women's and Children's Hospital, North Adelaide, 5006 South Australia, Australia.
| |
Collapse
|
38
|
Guo L, Zhou D, Pryse KM, Okunade AL, Su X. Fatty acid 2-hydroxylase mediates diffusional mobility of Raft-associated lipids, GLUT4 level, and lipogenesis in 3T3-L1 adipocytes. J Biol Chem 2010; 285:25438-47. [PMID: 20519515 PMCID: PMC2919107 DOI: 10.1074/jbc.m110.119933] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Revised: 05/26/2010] [Indexed: 01/27/2023] Open
Abstract
Straight chain fatty acid alpha-oxidation increases during differentiation of 3T3-L1 adipocytes, leading to a marked accumulation of odd chain length fatty acyl moieties. Potential roles of this pathway in adipocyte differentiation and lipogenesis are unknown. Mammalian fatty acid 2-hydroxylase (FA2H) was recently identified and suggested to catalyze the initial step of straight chain fatty acid alpha-oxidation. Accordingly, we examined whether FA2H modulates adipocyte differentiation and lipogenesis in mature adipocytes. FA2H level markedly increases during differentiation of 3T3-L1 adipocytes, and small interfering RNAs against FA2H inhibit the differentiation process. In mature adipocytes, depletion of FA2H inhibits basal and insulin-stimulated glucose uptake and lipogenesis, which are partially rescued by the enzymatic product of FA2H, 2-hydroxy palmitic acid. Expression of fatty-acid synthase and SCD1 was decreased in FA2H-depleted cells, and levels of GLUT4 and insulin receptor proteins were reduced. 2-Hydroxy fatty acids are enriched in cellular sphingolipids, which are components of membrane rafts. Accelerated diffusional mobility of raft-associated lipids was shown to enhance degradation of GLUT4 and insulin receptor in adipocytes. Consistent with this, depletion of FA2H appeared to increase raft lipid mobility as it significantly accelerated the rates of fluorescence recovery after photobleaching measurements of lipid rafts labeled with Alexa 488-conjugated cholera toxin subunit B. Moreover, the enhanced recovery rates were partially reversed by treatment with 2-hydroxy palmitic acid. In conclusion, our findings document the novel role of FA2H in adipocyte lipogenesis possibly by modulation of raft fluidity and level of GLUT4.
Collapse
Affiliation(s)
- Lin Guo
- From the Department of Internal Medicine, Center for Human Nutrition, and
| | - Dequan Zhou
- From the Department of Internal Medicine, Center for Human Nutrition, and
| | | | - Adewole L. Okunade
- From the Department of Internal Medicine, Center for Human Nutrition, and
| | - Xiong Su
- From the Department of Internal Medicine, Center for Human Nutrition, and
- Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
39
|
Ohno-Iwashita Y, Shimada Y, Hayashi M, Inomata M. Plasma membrane microdomains in aging and disease. Geriatr Gerontol Int 2010; 10 Suppl 1:S41-52. [DOI: 10.1111/j.1447-0594.2010.00600.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
40
|
Ande SR, Mishra S. Palmitoylation of prohibitin at cysteine 69 facilitates its membrane translocation and interaction with Eps 15 homology domain protein 2 (EHD2). Biochem Cell Biol 2010; 88:553-8. [DOI: 10.1139/o09-177] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Plasma membrane translocation of specific cytosolic proteins plays an important role in cell signaling pathways. We have recently shown that prohibitin (PHB) , a protein present in the plasma membranes of various cell types, interacts with Eps 15 homology domain protein 2 (EHD2), a lipid raft protein. However, the mechanism involved in membrane translocation of PHB is not known.We report that PHB undergoes palmitoylation at cysteine 69 (Cys69), and that this palmitoylation is required for PHB's membrane translocation. Furthermore, we demonstrate that membrane translocation of PHB facilitates tyrosine phosphorylation and its interaction with EHD2. Thus, the palmitoylation and membrane translocation of PHB and its interaction with EHD2 may play a role in cell signaling.
Collapse
Affiliation(s)
- Sudharsana Rao Ande
- Department of Internal Medicine, University of Manitoba, 843 John Buhler Research Centre, 715 McDermot Ave., Winnipeg, MB R3E 3P4, Canada
| | - Suresh Mishra
- Department of Internal Medicine, University of Manitoba, 843 John Buhler Research Centre, 715 McDermot Ave., Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
41
|
Hagen RM, Rodriguez-Cuenca S, Vidal-Puig A. An allostatic control of membrane lipid composition by SREBP1. FEBS Lett 2010; 584:2689-98. [PMID: 20385130 DOI: 10.1016/j.febslet.2010.04.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Accepted: 04/03/2010] [Indexed: 12/19/2022]
Abstract
The maintenance of membrane lipid composition within strict limits is critical to maintain optimum cellular function. The biophysical properties of the membrane can be influenced among other factors by the saturation/unsaturation of the phospholipid fatty acyl chain. The rate-limiting enzyme in unsaturated fatty acid biosynthesis is the desaturase enzyme which in turn is regulated by the lipid transcription factor sterol regulatory element binding protein (SREBP1). In this review, we collect some evidence suggesting SREBP1 network as an important allostatic regulator necessary to maintain the pool of unsaturated fatty acid lipid species that can be incorporated into biological membranes.
Collapse
Affiliation(s)
- Rachel M Hagen
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, UK.
| | | | | |
Collapse
|
42
|
Yi P, Nguyên DT, Higa-Nishiyama A, Auguste P, Bouchecareilh M, Dominguez M, Bielmann R, Palcy S, Liu JF, Chevet E. MAPK scaffolding by BIT1 in the Golgi complex modulates stress resistance. J Cell Sci 2010; 123:1060-72. [PMID: 20197408 DOI: 10.1242/jcs.059717] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The endoplasmic reticulum (ER) is an essential organelle whose major functions are to ensure proper secretory protein folding and trafficking. These mechanisms involve the activation of specific ER-resident molecular machines, which might be regulated by their membranous environments. Based on this observation, we aimed to characterize the proteome of ER-membrane microdomains to identify new components of the ER that have a role in secretory pathway-associated functions. Using this approach with dog pancreatic rough microsomes, we found that mitochondrial Bcl-2 inhibitor of transcription (BIT1) localized in the early secretory pathway and accumulated in the Golgi complex. Using both a chimeric protein of the luminal and transmembrane domains of ER-resident TRAPalpha and the cytosolic domain of BIT1, and silencing of BIT1 expression, we perturbed endogenous BIT1 oligomerization and localization to the Golgi. This led to enhanced ERK signaling from the Golgi complex, which resulted in improved stress resistance. This work provides the first evidence for the existence of ER microdomains that are involved in the regulation of BIT1 structure and trafficking, and identifies BIT1 as a negative regulator of the ERK-MAPK signaling pathway in the Golgi.
Collapse
Affiliation(s)
- Ping Yi
- Avenir, INSERM U889, Bordeaux, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Blouin CM, Prado C, Takane KK, Lasnier F, Garcia-Ocana A, Ferré P, Dugail I, Hajduch E. Plasma membrane subdomain compartmentalization contributes to distinct mechanisms of ceramide action on insulin signaling. Diabetes 2010; 59:600-10. [PMID: 19959757 PMCID: PMC2828662 DOI: 10.2337/db09-0897] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Ceramide is now recognized as a negative regulator of insulin signaling by impairing protein kinase B (PKB)/Akt activation. In different cells, two distinct mechanisms have been proposed to mediate ceramide inhibition of PKB/Akt: one involving atypical protein kinase C zeta (PKCzeta) and the other the protein phosphatase-2 (PP2A). We hypothesized that ceramide action through PKCzeta or PP2A might depend on plasma membrane (PM) structural organization and especially on caveolin-enriched domain (CEM) abundance. RESEARCH DESIGN AND METHODS We have used different PKCzeta mutant constructs or the PP2A inhibitor, okadaic acid (OKA), to selectively inhibit PKCzeta- and PP2A-dependent pathways in cells expressing different caveolin-1 levels and evaluated the impact of insulin and ceramide on PKB/Akt activity in different PM subdomains. RESULTS Although the PKCzeta-mediated negative effect of ceramide on insulin-stimulated PKB/Akt was dominant in adipocytes, a ceramide action through PP2A outside CEMs, prevented by OKA, was also unraveled. To test the importance of CEM to direct ceramide action through the PKCzeta pathway, we treated 3T3-L1 preadipocytes devoid of CEMs with ceramide and we saw a shift of the lipid-negative action on PKB/Akt to a PP2A-mediated mechanism. In fibroblasts with low CEM abundance, the ceramide-activated PP2A pathway dominated, but could be shifted to a ceramide-activated PKCzeta pathway after caveolin-1 overexpression. CONCLUSIONS Our results show that ceramide can switch from a PKCzeta-dependent mechanism to a PP2A pathway, acting negatively on PKB/Akt, and hence revealing a critical role of CEMs of the PM in this process.
Collapse
Affiliation(s)
- Cédric M. Blouin
- Centre de Recherche des Cordeliers, INSERM, UMR-S 872, Paris, France
- Université Pierre et Marie Curie–Paris 6, UMR-S 872, Paris, France
- Université Paris Descartes, UMR-S 872, Paris, France
| | - Cécilia Prado
- Centre de Recherche des Cordeliers, INSERM, UMR-S 872, Paris, France
- Université Pierre et Marie Curie–Paris 6, UMR-S 872, Paris, France
- Université Paris Descartes, UMR-S 872, Paris, France
| | - Karen K. Takane
- Division of Endocrinology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Françoise Lasnier
- Centre de Recherche des Cordeliers, INSERM, UMR-S 872, Paris, France
- Université Pierre et Marie Curie–Paris 6, UMR-S 872, Paris, France
- Université Paris Descartes, UMR-S 872, Paris, France
| | - Adolfo Garcia-Ocana
- Division of Endocrinology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Pascal Ferré
- Centre de Recherche des Cordeliers, INSERM, UMR-S 872, Paris, France
- Université Pierre et Marie Curie–Paris 6, UMR-S 872, Paris, France
- Université Paris Descartes, UMR-S 872, Paris, France
| | - Isabelle Dugail
- Centre de Recherche des Cordeliers, INSERM, UMR-S 872, Paris, France
- Université Pierre et Marie Curie–Paris 6, UMR-S 872, Paris, France
- Université Paris Descartes, UMR-S 872, Paris, France
| | - Eric Hajduch
- Centre de Recherche des Cordeliers, INSERM, UMR-S 872, Paris, France
- Université Pierre et Marie Curie–Paris 6, UMR-S 872, Paris, France
- Université Paris Descartes, UMR-S 872, Paris, France
- Corresponding author: Eric Hajduch,
| |
Collapse
|
44
|
Sánchez-Wandelmer J, Dávalos A, de la Peña G, Cano S, Giera M, Canfrán-Duque A, Bracher F, Martín-Hidalgo A, Fernández-Hernando C, Lasunción MA, Busto R. Haloperidol disrupts lipid rafts and impairs insulin signaling in SH-SY5Y cells. Neuroscience 2010; 167:143-53. [PMID: 20123000 DOI: 10.1016/j.neuroscience.2010.01.051] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Revised: 01/13/2010] [Accepted: 01/25/2010] [Indexed: 11/17/2022]
Abstract
Haloperidol exerts its therapeutic effects basically by acting on dopamine receptors. We previously reported that haloperidol inhibits cholesterol biosynthesis in cultured cells. In the present work we investigated its effects on lipid-raft composition and functionality. In both neuroblastoma SH-SY5Y and promyelocytic HL-60 human cell lines, haloperidol inhibited cholesterol biosynthesis resulting in a decrease of the cell cholesterol content and the accumulation of different sterol intermediates (7-dehydrocholesterol, zymostenol and cholesta-8,14-dien-3beta-ol) depending on the dose of the drug. As a consequence, the cholesterol content in lipid rafts was greatly reduced, and several pre-cholesterol sterols, particularly cholesta-8,14-dien-3beta-ol, were incorporated into the cell membrane. This was accompanied by the disruption of lipid rafts, with redistribution of flotillin-1 and Fyn and the impairment of insulin-Akt signaling. Supplementing the medium with free cholesterol abrogated the effects of haloperidol on lipid-raft composition and functionality. LDL (low-density lipoprotein), a physiological vehicle of cholesterol in plasma, was much less effective in preventing the effects of haloperidol, which is attributed to the drug's inhibition of intracellular vesicular trafficking. These effects on cellular cholesterol homeostasis that ultimately result in the alteration of lipid-raft-dependent insulin signaling action may underlie some of the metabolic effects of this widely used antipsychotic.
Collapse
|
45
|
Inokuchi JI. Membrane microdomains and insulin resistance. FEBS Lett 2009; 584:1864-71. [PMID: 19822143 DOI: 10.1016/j.febslet.2009.10.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Revised: 10/03/2009] [Accepted: 10/06/2009] [Indexed: 11/18/2022]
Abstract
A new concept, that "metabolic disorders, such as type 2 diabetes, are membrane microdomain disorders caused by aberrant expression of gangliosides", has arisen. By examining this working hypothesis, we demonstrate the molecular pathogenesis of type 2 diabetes and insulin resistance focusing on the interaction between insulin receptor and gangliosides in microdomains and propose the new therapeutic strategy "membrane microdomain ortho-signaling therapy".
Collapse
Affiliation(s)
- Jin-ichi Inokuchi
- Division of Glycopathology, Institute of Molecular Biomembranes and Glycobiology, Tohoku Pharmaceutical University, Aoba-ku, Sendai, Miyagi, Japan.
| |
Collapse
|
46
|
Taghibiglou C, Bradley CA, Gaertner T, Li Y, Wang Y, Wang YT. Mechanisms involved in cholesterol-induced neuronal insulin resistance. Neuropharmacology 2009; 57:268-76. [DOI: 10.1016/j.neuropharm.2009.05.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2008] [Revised: 04/05/2009] [Accepted: 05/26/2009] [Indexed: 12/27/2022]
|
47
|
Insulin induced phosphorylation of prohibitin at tyrosine 114 recruits Shp1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:1372-8. [PMID: 19497338 DOI: 10.1016/j.bbamcr.2009.05.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2009] [Revised: 05/14/2009] [Accepted: 05/26/2009] [Indexed: 12/22/2022]
Abstract
Prohibitin (PHB or PHB1) is an evolutionarily conserved ubiquitously expressed multifunctional protein and is present in various cellular compartments. Phosphorylation of PHB has been suggested as one of the potential mechanisms in the regulation of its various functions however exact sites of phosphorylation remain to be determined. To better understand the functional relevance of phosphorylation of PHB, we have explored the potential sites of phosphorylation using combination of approaches including phosphoamino specific immunoblotting, proteolysis, two-dimensional gel electrophoresis, phosphoamino acid analysis and site-directed mutagenesis techniques and report that tyrosine 114 (Tyr 114) in PHB is phosphorylated in response to insulin stimulation. In addition, using active insulin receptor (IR) and synthetic biotinylated PHB peptide (PHB(107-121)) we have shown that IR also phosphorylates Tyr 114 in an in vitro kinase assay. Phosphorylation of PHB at Tyr 114 was confirmed by immunoblotting using anti-phosphoTyr 114 specific antibody. Furthermore, we demonstrate that SH2 domain containing tyrosine phosphatase-1 (Shp1) co-immunoprecipitate with PHB antiserum after insulin induced phosphorylation of PHB. Biotinylated-PHB(107-121) peptide phosphorylated at Tyr 114 was also able to pull down Shp1 in pull down assays. Non-phosphorylated PHB(107-121) peptide, corresponding PHB2(121-135) peptide and Tyr114Phe mutant-PHB fail to pull down Shp1. In summary, we have identified Tyr 114 in PHB as an important site of phosphorylation and phosphorylation at this residue creates a binding site for Shp1 both in vivo and in vitro.
Collapse
|
48
|
Bazuine M, Stenkula KG, Cam M, Arroyo M, Cushman SW. Guardian of corpulence: a hypothesis on p53 signaling in the fat cell. ACTA ACUST UNITED AC 2009; 4:231-243. [PMID: 20126301 DOI: 10.2217/clp.09.2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Adipocytes provide an organism with fuel in times of caloric deficit, and are an important type of endocrine cell in the maintenance of metabolic homeostasis. In addition, as a lipid-sink, adipocytes serve an equally important role in the protection of organs from the damaging effects of ectopic lipid deposition. For the organism, it is of vital importance to maintain adipocyte viability, yet the fat depot is a demanding extracellular environment with high levels of interstitial free fatty acids and associated lipotoxic effects. These surroundings are less than beneficial for the overall health of any resident cell, adipocyte and preadipocyte alike. In this review, we discuss the process of adipogenesis and the potential involvement of the p53 tumor-suppressor protein in alleviating some of the cellular stress experienced by these cells. In particular, we discuss p53-mediated mechanisms that prevent damage caused by reactive oxygen species and the effects of lipotoxicity. We also suggest the potential for two p53 target genes, START domain-containing protein 4 (StARD4) and oxysterol-binding protein (OSBP), with the concomitant synthesis of the signaling molecule oxysterol, to participate in adipogenesis.
Collapse
Affiliation(s)
- Merlijn Bazuine
- Experimental Diabetes, Metabolism & Nutrition Section, Diabetes Branch, NIDDK, NIH, Building 10-CRC, Room 5W-5816, 10 Center Drive, Bethesda, MD 20892, USA, Tel.: +1 301 496 7354, ,
| | | | | | | | | |
Collapse
|
49
|
Lalazar G, Ben Ya'acov A, Livovsky DM, El Haj M, Pappo O, Preston S, Zolotarov L, Ilan Y. Beta-glycoglycosphingolipid-induced alterations of the STAT signaling pathways are dependent on CD1d and the lipid raft protein flotillin-2. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:1390-9. [PMID: 19246642 DOI: 10.2353/ajpath.2009.080841] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Beta-glucosylceramide has been shown to affect natural killer T cell function in models of inflammation. We, therefore, investigated the effects of different beta-glycosphingolipids, including beta-glucosylceramide, on STAT (signal transducers and activators of transcription) signaling pathways and determined whether these effects were mediated by lipid raft microdomains and/or CD1d molecules. The effects of alpha- and beta-structured ligands on the lipid raft protein flotillin-2 were studied in both natural killer T hybridoma cells and leptin-deficient mice. To determine whether CD1d was involved in the effects of the beta-glycosphingolipids, an anti-CD1d blocking antibody was used in a cell proliferation assay system. The downstream effects on the protein phosphorylation levels of STAT1, STAT3, and STAT6 were examined in both immune-mediated hepatitis and hepatoma models. The effects of beta-glycosphingolipids on the STAT signaling pathways were found to be dependent on CD1d. Lipid rafts were affected by both the dose and ratio of the beta-glycosphingolipids and the acyl chain length, and these effects were followed by downstream effects on STAT proteins. Our results show that beta-glycosphingolipids have beneficial effects in natural killer T cell-dependent immune-mediated metabolic and malignant animal models in vivo.
Collapse
Affiliation(s)
- Gadi Lalazar
- Liver Unit, Department of Medicine, Hebrew University-Hadassah Medical Center, P.O.B 12000, Jerusalem, Israel, IL-91120
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Ande SR, Moulik S, Mishra S. Interaction between O-GlcNAc modification and tyrosine phosphorylation of prohibitin: implication for a novel binary switch. PLoS One 2009; 4:e4586. [PMID: 19238206 PMCID: PMC2642629 DOI: 10.1371/journal.pone.0004586] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2008] [Accepted: 01/07/2009] [Indexed: 11/20/2022] Open
Abstract
Prohibitin (PHB or PHB1) is an evolutionarily conserved, multifunctional protein which is present in various cellular compartments including the plasma membrane. However, mechanisms involved in various functions of PHB are not fully explored yet. Here we report for the first time that PHB interacts with O-linked β-N-acetylglucosamine transferase (O-GlcNAc transferase, OGT) and is O-GlcNAc modified; and also undergoes tyrosine phosphorylation in response to insulin. Tyrosine 114 (Tyr114) and tyrosine 259 (Tyr259) in PHB are in the close proximity of potential O-GlcNAc sites serine 121 (Ser121) and threonine 258 (Thr258) respectively. Substitution of Tyr114 and Tyr259 residues in PHB with phenylalanine by site-directed mutagenesis results in reduced tyrosine phosphorylation as well as reduced O-GlcNAc modification of PHB. Surprisingly, this also resulted in enhanced tyrosine phosphorylation and activity of OGT. This is attributed to the presence of similar tyrosine motifs in PHB and OGT. Substitution of Ser121 and Thr258 with alanine and isoleucine respectively resulted in attenuation of O-GlcNAc modification and increased tyrosine phosphorylation of PHB suggesting an association between these two dynamic modifications. Sequence analysis of O-GlcNAc modified proteins having known O-GlcNAc modification site(s) or known tyrosine phosphorylation site(s) revealed a strong potential association between these two posttranslational modifications in various proteins. We speculate that O-GlcNAc modification and tyrosine phosphorylation of PHB play an important role in tyrosine kinase signaling pathways including insulin, growth factors and immune receptors signaling. In addition, we propose that O-GlcNAc modification and tyrosine phosphorylation is a novel previously unidentified binary switch which may provide new mechanistic insights into cell signaling pathways and is open for direct experimental examination.
Collapse
Affiliation(s)
- Sudharsana R Ande
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | |
Collapse
|