1
|
SantaCruz-Calvo S, Saraswat S, Kalantar GH, Zukowski E, Marszalkowski H, Javidan A, Gholamrezaeinejad F, Bharath LP, Kern PA, Zhang XD, Nikolajczyk BS. A unique inflammaging profile generated by T cells from people with obesity is metformin resistant. GeroScience 2024:10.1007/s11357-024-01441-4. [PMID: 39708215 DOI: 10.1007/s11357-024-01441-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 11/07/2024] [Indexed: 12/23/2024] Open
Abstract
The alarmingly high prevalence of obesity in older adults coupled with the negative health effects of chronic inflammation in both obesity and aging highlight the importance of studies investigating the impacts of obesity on age-related inflammation. Since shifts in peripheral T-cell metabolism and function drive systemic inflammation in both obesity and aging, we hypothesize that obesity impacts the Th17-dominated inflammaging profile we identified in lean subjects and thus modifies the anti-inflammatory effects of geroprotective drugs like metformin. New cytokine profiling data showed that CD4+ T cells from older people with obesity generate a profile that specifically excludes Th17 cytokines. Metformin failed to change the age-associated T-cell profile in obesity, despite lowering both mitochondrial respiration and reactive oxygen species (ROS) production. Metformin did not improve macroautophagy in T cells from older people with obesity, in sharp contrast to the ability of metformin to promote autophagy in T cells from older lean subjects. These data indicate that body mass index modifies the mechanisms supporting inflammaging in T cells from older subjects, and that metformin-mediated restoration of redox balance is insufficient to stem obesity-associated inflammaging. We conclude that obesity fundamentally changes the mechanisms that promote inflammaging, and thus obesity becomes a critical consideration for clinical trials of geroprotective agents such as metformin.
Collapse
Affiliation(s)
- S SantaCruz-Calvo
- Department of Pharmacology & Nutritional Sciences, Diabetes and Obesity Research Priority Area, University of Kentucky, Lexington, KY, USA
| | - S Saraswat
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - G H Kalantar
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - E Zukowski
- Department of Nutrition & Public Health, Merrimack College, North Andover, MA, USA
| | - H Marszalkowski
- Department of Biology, Merrimack College, North Andover, MA, USA
| | - A Javidan
- Department of Pharmacology & Nutritional Sciences, Diabetes and Obesity Research Priority Area, University of Kentucky, Lexington, KY, USA
| | - F Gholamrezaeinejad
- Department of Pharmacology & Nutritional Sciences, Diabetes and Obesity Research Priority Area, University of Kentucky, Lexington, KY, USA
| | - L P Bharath
- Department of Nutrition & Public Health, Merrimack College, North Andover, MA, USA
| | - P A Kern
- Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
- Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, KY, USA
| | - X D Zhang
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA
- Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, KY, USA
| | - B S Nikolajczyk
- Department of Pharmacology & Nutritional Sciences, Diabetes and Obesity Research Priority Area, University of Kentucky, Lexington, KY, USA.
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, KY, USA.
- Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
2
|
Rockhold JD, Marszalkowski H, Sannella M, Gibney K, Murphy L, Zukowski E, Kalantar GH, SantaCruz-Calvo S, Hart SN, Kuhn MK, Yu J, Stefanik O, Chase G, Proctor EA, Hasturk H, Nikolajczyk BS, Bharath LP. Everolimus alleviates CD4 + T cell inflammation by regulating autophagy and cellular redox homeostasis. GeroScience 2024; 46:5681-5699. [PMID: 38761287 PMCID: PMC11493941 DOI: 10.1007/s11357-024-01187-z] [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: 10/10/2023] [Accepted: 04/30/2024] [Indexed: 05/20/2024] Open
Abstract
Aging is associated with the onset and progression of multiple diseases, which limit health span. Chronic low-grade inflammation in the absence of overt infection is considered the simmering source that triggers age-associated diseases. Failure of many cellular processes during aging is mechanistically linked to inflammation; however, the overall decline in the cellular homeostasis mechanism of autophagy has emerged as one of the top and significant inducers of inflammation during aging, frequently known as inflammaging. Thus, physiological or pharmacological interventions aimed at improving autophagy are considered geroprotective. Rapamycin analogs (rapalogs) are known for their ability to inhibit mTOR and thus regulate autophagy. This study assessed the efficacy of everolimus, a rapalog, in regulating inflammatory cytokine production in T cells from older adults. CD4+ T cells from older adults were treated with a physiological dose of everolimus (0.01 µM), and indices of autophagy and inflammation were assessed to gain a mechanistic understanding of the effect of everolimus on inflammation. Everolimus (Ever) upregulated autophagy and broadly alleviated inflammatory cytokines produced by multiple T cell subsets. Everolimus's ability to alleviate the cytokines produced by Th17 subsets of T cells, such as IL-17A and IL-17F, was dependent on autophagy and antioxidant signaling pathways. Repurposing the antineoplastic drug everolimus for curbing inflammaging is promising, given the drug's ability to restore multiple cellular homeostasis mechanisms.
Collapse
Affiliation(s)
- Jack Donato Rockhold
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA
| | | | - Marco Sannella
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA
| | - Kaleigh Gibney
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA
| | - Lyanne Murphy
- Department of Biology, Merrimack College, North Andover, MA, USA
| | - Emelia Zukowski
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA
| | - Gabriella H Kalantar
- Dept of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - Sara SantaCruz-Calvo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
- Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA
| | - Samantha N Hart
- Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Madison K Kuhn
- Department of Neurosurgery, Pharmacology, and Biomedical Engineering and Center for Neural Engineering, Pennsylvania State University, Hershey, PA, USA
| | - Jingting Yu
- Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Olivia Stefanik
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA
| | - Gabrielle Chase
- Department of Chemistry and Biochemistry, Merrimack College, North Andover, MA, USA
| | - Elizabeth A Proctor
- Department of Neurosurgery, Pharmacology, and Biomedical Engineering and Center for Neural Engineering, Pennsylvania State University, Hershey, PA, USA
- Department of Engineering Science & Mechanics, Pennsylvania State University, University Park, PA, USA
| | | | - Barbara S Nikolajczyk
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
- Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA
| | - Leena P Bharath
- Department of Health Sciences and Nutrition, Merrimack College, North Andover, MA, USA.
| |
Collapse
|
3
|
Chiu PL, Orjuela JD, de Groot BL, Aponte-Santamaría C, Walz T. Structure and dynamics of cholesterol-mediated aquaporin-0 arrays and implications for lipid rafts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.16.540959. [PMID: 37292626 PMCID: PMC10245776 DOI: 10.1101/2023.05.16.540959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Aquaporin-0 (AQP0) tetramers form square arrays in lens membranes through a yet unknown mechanism, but lens membranes are enriched in sphingomyelin and cholesterol. Here, we determined electron crystallographic structures of AQP0 in sphingomyelin/cholesterol membranes and performed molecular dynamics (MD) simulations to establish that the observed cholesterol positions represent those seen around an isolated AQP0 tetramer and that the AQP0 tetramer largely defines the location and orientation of most of its associated cholesterol molecules. At a high concentration, cholesterol increases the hydrophobic thickness of the annular lipid shell around AQP0 tetramers, which may thus cluster to mitigate the resulting hydrophobic mismatch. Moreover, neighboring AQP0 tetramers sandwich a cholesterol deep in the center of the membrane. MD simulations show that the association of two AQP0 tetramers is necessary to maintain the deep cholesterol in its position and that the deep cholesterol increases the force required to laterally detach two AQP0 tetramers, not only due to protein-protein contacts but also due to increased lipid-protein complementarity. Since each tetramer interacts with four such 'glue' cholesterols, avidity effects may stabilize larger arrays. The principles proposed to drive AQP0 array formation could also underlie protein clustering in lipid rafts.
Collapse
|
4
|
Conway R, Rockhold JD, SantaCruz-Calvo S, Zukowski E, Pugh GH, Hasturk H, Kern PA, Nikolajczyk BS, Bharath LP. Obesity and Fatty Acids Promote Mitochondrial Translocation of STAT3 Through ROS-Dependent Mechanisms. FRONTIERS IN AGING 2022; 3:924003. [PMID: 35928250 PMCID: PMC9344057 DOI: 10.3389/fragi.2022.924003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/08/2022] [Indexed: 11/29/2022]
Abstract
Obesity promotes the onset and progression of metabolic and inflammatory diseases such as type 2 diabetes. The chronic low-grade inflammation that occurs during obesity triggers multiple signaling mechanisms that negatively affect organismal health. One such mechanism is the persistent activation and mitochondrial translocation of STAT3, which is implicated in inflammatory pathologies and many types of cancers. STAT3 in the mitochondria (mitoSTAT3) alters electron transport chain activity, thereby influencing nutrient metabolism and immune response. PBMCs and CD4+ T cells from obese but normal glucose-tolerant (NGT) middle-aged subjects had higher phosphorylation of STAT3 on residue serine 727 and more mitochondrial accumulation of STAT3 than cells from lean subjects. To evaluate if circulating lipid overabundance in obesity is responsible for age- and sex-matched mitoSTAT3, cells from lean subjects were challenged with physiologically relevant doses of the saturated and monounsaturated fatty acids, palmitate and oleate, respectively. Fatty acid treatment caused robust accumulation of mitoSTAT3 in all cell types, which was independent of palmitate-induced impairments in autophagy. Co-treatment of cells with fatty acid and trehalose prevented STAT3 phosphorylation and mitochondrial accumulation in an autophagy-independent but cellular peroxide-dependent mechanism. Pharmacological blockade of mitoSTAT3 either by a mitochondria-targeted STAT3 inhibitor or ROS scavenging prevented obesity and fatty acid-induced production of proinflammatory cytokines IL-17A and IL-6, thus establishing a mechanistic link between mitoSTAT3 and inflammatory cytokine production.
Collapse
Affiliation(s)
- Rachel Conway
- Department of Nutrition and Public Health, Merrimack College, North Andover, MA, United States
| | - Jack Donato Rockhold
- Department of Nutrition and Public Health, Merrimack College, North Andover, MA, United States
| | - Sara SantaCruz-Calvo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
- Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, United States
| | - Emelia Zukowski
- Department of Nutrition and Public Health, Merrimack College, North Andover, MA, United States
| | - Gabriella H. Pugh
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, United States
| | | | - Philip A. Kern
- Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, United States
- Department of Medicine, University of Kentucky, Lexington, KY, United States
| | - Barbara S. Nikolajczyk
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
- Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, United States
| | - Leena P. Bharath
- Department of Nutrition and Public Health, Merrimack College, North Andover, MA, United States
| |
Collapse
|
5
|
Sebestyén V, Nagy É, Mocsár G, Volkó J, Szilágyi O, Kenesei Á, Panyi G, Tóth K, Hajdu P, Vámosi G. Role of C-Terminal Domain and Membrane Potential in the Mobility of Kv1.3 Channels in Immune Synapse Forming T Cells. Int J Mol Sci 2022; 23:ijms23063313. [PMID: 35328733 PMCID: PMC8952507 DOI: 10.3390/ijms23063313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Voltage-gated Kv1.3 potassium channels are essential for maintaining negative membrane potential during T-cell activation. They interact with membrane-associated guanylate kinases (MAGUK-s) via their C-terminus and with TCR/CD3, leading to enrichment at the immunological synapse (IS). Molecular interactions and mobility may impact each other and the function of these proteins. We aimed to identify molecular determinants of Kv1.3 mobility, applying fluorescence correlation spectroscopy on human Jurkat T-cells expressing WT, C-terminally truncated (ΔC), and non-conducting mutants of mGFP-Kv1.3. ΔC cannot interact with MAGUK-s and is not enriched at the IS, whereas cells expressing the non-conducting mutant are depolarized. Here, we found that in standalone cells, mobility of ΔC increased relative to the WT, likely due to abrogation of interactions, whereas mobility of the non-conducting mutant decreased, similar to our previous observations on other membrane proteins in depolarized cells. At the IS formed with Raji B-cells, mobility of WT and non-conducting channels, unlike ΔC, was lower than outside the IS. The Kv1.3 variants possessing an intact C-terminus had lower mobility in standalone cells than in IS-engaged cells. This may be related to the observed segregation of F-actin into a ring-like structure at the periphery of the IS, leaving much of the cell almost void of F-actin. Upon depolarizing treatment, mobility of WT and ΔC channels decreased both in standalone and IS-engaged cells, contrary to non-conducting channels, which themselves caused depolarization. Our results support that Kv1.3 is enriched at the IS via its C-terminal region regardless of conductivity, and that depolarization decreases channel mobility.
Collapse
Affiliation(s)
- Veronika Sebestyén
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (V.S.); (É.N.); (G.M.); (J.V.); (O.S.); (Á.K.); (G.P.); (K.T.)
| | - Éva Nagy
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (V.S.); (É.N.); (G.M.); (J.V.); (O.S.); (Á.K.); (G.P.); (K.T.)
| | - Gábor Mocsár
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (V.S.); (É.N.); (G.M.); (J.V.); (O.S.); (Á.K.); (G.P.); (K.T.)
| | - Julianna Volkó
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (V.S.); (É.N.); (G.M.); (J.V.); (O.S.); (Á.K.); (G.P.); (K.T.)
| | - Orsolya Szilágyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (V.S.); (É.N.); (G.M.); (J.V.); (O.S.); (Á.K.); (G.P.); (K.T.)
| | - Ádám Kenesei
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (V.S.); (É.N.); (G.M.); (J.V.); (O.S.); (Á.K.); (G.P.); (K.T.)
| | - György Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (V.S.); (É.N.); (G.M.); (J.V.); (O.S.); (Á.K.); (G.P.); (K.T.)
| | - Katalin Tóth
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (V.S.); (É.N.); (G.M.); (J.V.); (O.S.); (Á.K.); (G.P.); (K.T.)
- Division Biophysics of Macromolecules, German Cancer Research Center, D-69120 Heidelberg, Germany
| | - Péter Hajdu
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (V.S.); (É.N.); (G.M.); (J.V.); (O.S.); (Á.K.); (G.P.); (K.T.)
- Department of Biophysics and Cell Biology, Faculty of Dentistry, University of Debrecen, H-4032 Debrecen, Hungary
- Correspondence: (P.H.); (G.V.)
| | - György Vámosi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (V.S.); (É.N.); (G.M.); (J.V.); (O.S.); (Á.K.); (G.P.); (K.T.)
- Correspondence: (P.H.); (G.V.)
| |
Collapse
|
6
|
Charpentier JC, King PD. Mechanisms and functions of endocytosis in T cells. Cell Commun Signal 2021; 19:92. [PMID: 34503523 PMCID: PMC8427877 DOI: 10.1186/s12964-021-00766-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/17/2021] [Indexed: 11/11/2022] Open
Abstract
Once thought of primarily as a means to neutralize pathogens or to facilitate feeding, endocytosis is now known to regulate a wide range of eukaryotic cell processes. Among these are regulation of signal transduction, mitosis, lipid homeostasis, and directed migration, among others. Less well-appreciated are the roles various forms of endocytosis plays in regulating αβ and, especially, γδ T cell functions, such as T cell receptor signaling, antigen discovery by trogocytosis, and activated cell growth. Herein we examine the contribution of both clathrin-mediated and clathrin-independent mechanisms of endocytosis to T cell biology. Video Abstract
Collapse
Affiliation(s)
- John C Charpentier
- Department of Microbiology and Immunology, University of Michigan Medical School, 6606 Med Sci II, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5620, USA
| | - Philip D King
- Department of Microbiology and Immunology, University of Michigan Medical School, 6606 Med Sci II, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5620, USA.
| |
Collapse
|
7
|
Thermogenic Activation Downregulates High Mitophagy Rate in Human Masked and Mature Beige Adipocytes. Int J Mol Sci 2020; 21:ijms21186640. [PMID: 32927882 PMCID: PMC7555361 DOI: 10.3390/ijms21186640] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/07/2020] [Accepted: 09/07/2020] [Indexed: 12/16/2022] Open
Abstract
Thermogenic brown and beige adipocytes oxidize metabolic substrates producing heat, mainly by the mitochondrial uncoupling protein UCP1, and can thus counteract obesity. Masked beige adipocytes possess white adipocyte-like morphology, but can be made thermogenic by adrenergic stimuli. We investigated the regulation of mitophagy upon thermogenic activation of human masked and mature beige adipocytes. Human primary abdominal subcutaneous adipose-derived stromal cells (hASCs) and Simpson-Golabi-Behmel syndrome (SGBS) preadipocytes were differentiated to white and beige adipocytes, then their cAMP-induced thermogenic potential was assessed by detecting increased expressions of UCP1, mitochondrial DNA content and respiratory chain complex subunits. cAMP increased the thermogenic potential of white adipocytes similarly to beige ones, indicating the presence of a masked beige population. In unstimulated conditions, a high autophagic flux and mitophagy rates (demonstrated by LC3 punctae and TOM20 co-immunostaining) were observed in white adipocytes, while these were lower in beige adipocytes. Silencing and gene expression experiments showed that the ongoing mitophagy was Parkin-independent. cAMP treatment led to the downregulation of mitophagy through PKA in both types of adipocytes, resulting in more fragmented mitochondria and increased UCP1 levels. Our data indicates that mitophagy is repressed upon encountering a short-term adrenergic stimulus, as a fast regulatory mechanism to provide high mitochondrial content for thermogenesis.
Collapse
|
8
|
Bharath LP, Agrawal M, McCambridge G, Nicholas DA, Hasturk H, Liu J, Jiang K, Liu R, Guo Z, Deeney J, Apovian CM, Snyder-Cappione J, Hawk GS, Fleeman RM, Pihl RMF, Thompson K, Belkina AC, Cui L, Proctor EA, Kern PA, Nikolajczyk BS. Metformin Enhances Autophagy and Normalizes Mitochondrial Function to Alleviate Aging-Associated Inflammation. Cell Metab 2020; 32:44-55.e6. [PMID: 32402267 PMCID: PMC7217133 DOI: 10.1016/j.cmet.2020.04.015] [Citation(s) in RCA: 392] [Impact Index Per Article: 78.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 01/28/2020] [Accepted: 04/20/2020] [Indexed: 12/20/2022]
Abstract
Age is a non-modifiable risk factor for the inflammation that underlies age-associated diseases; thus, anti-inflammaging drugs hold promise for increasing health span. Cytokine profiling and bioinformatic analyses showed that Th17 cytokine production differentiates CD4+ T cells from lean, normoglycemic older and younger subjects, and mimics a diabetes-associated Th17 profile. T cells from older compared to younger subjects also had defects in autophagy and mitochondrial bioenergetics that associate with redox imbalance. Metformin ameliorated the Th17 inflammaging profile by increasing autophagy and improving mitochondrial bioenergetics. By contrast, autophagy-targeting siRNA disrupted redox balance in T cells from young subjects and activated the Th17 profile by activating the Th17 master regulator, STAT3, which in turn bound IL-17A and F promoters. Mitophagy-targeting siRNA failed to activate the Th17 profile. We conclude that metformin improves autophagy and mitochondrial function largely in parallel to ameliorate a newly defined inflammaging profile that echoes inflammation in diabetes.
Collapse
Affiliation(s)
- Leena P Bharath
- Department of Nutrition and Public Health, Merrimack College, North Andover, MA, USA
| | - Madhur Agrawal
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA; Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA
| | - Grace McCambridge
- Department of Nutrition and Public Health, Merrimack College, North Andover, MA, USA
| | - Dequina A Nicholas
- Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of California, San Diego, San Diego, CA, USA
| | | | - Jing Liu
- Department of Computer Science, University of Kentucky, Lexington, KY, USA
| | - Kai Jiang
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Rui Liu
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| | - Zhenheng Guo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Jude Deeney
- Department of Medicine, Endocrinology, Diabetes & Nutrition, Boston University School of Medicine, Boston, MA, USA
| | - Caroline M Apovian
- Department of Medicine, Endocrinology, Diabetes & Nutrition, Boston University School of Medicine, Boston, MA, USA
| | - Jennifer Snyder-Cappione
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA; Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA
| | - Gregory S Hawk
- Department of Statistics, University of Kentucky, Lexington, KY, USA
| | - Rebecca M Fleeman
- Departments of Neurosurgery and Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Riley M F Pihl
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA
| | | | - Anna C Belkina
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Licong Cui
- Department of Computer Science, University of Kentucky, Lexington, KY, USA; School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Elizabeth A Proctor
- Departments of Neurosurgery and Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA; Departments of Biomedical Engineering, and Engineering Science & Mechanics and Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
| | - Philip A Kern
- Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA; Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Barbara S Nikolajczyk
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA; Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
9
|
Abstract
Apolipoprotein A-I binding protein (AIBP) is a recently identified innate anti-inflammatory factor. Here, we show that AIBP inhibited HIV replication by targeting lipid rafts and reducing virus-cell fusion. Importantly, AIBP selectively reduced levels of rafts on cells stimulated by an inflammatory stimulus or treated with extracellular vesicles containing HIV-1 protein Nef without affecting rafts on nonactivated cells. Accordingly, fusion of monocyte-derived macrophages with HIV was sensitive to AIBP only in the presence of Nef. Silencing of endogenous AIBP significantly upregulated HIV-1 replication. Interestingly, HIV-1 replication in cells from donors with the HLA-B*35 genotype, associated with rapid progression of HIV disease, was not inhibited by AIBP. These results suggest that AIBP is an innate anti-HIV factor that targets virus-cell fusion. Apolipoprotein A-I binding protein (AIBP) is a protein involved in regulation of lipid rafts and cholesterol efflux. AIBP has been suggested to function as a protective factor under several sets of pathological conditions associated with increased abundance of lipid rafts, such as atherosclerosis and acute lung injury. Here, we show that exogenously added AIBP reduced the abundance of lipid rafts and inhibited HIV replication in vitro as well as in HIV-infected humanized mice, whereas knockdown of endogenous AIBP increased HIV replication. Endogenous AIBP was much more abundant in activated T cells than in monocyte-derived macrophages (MDMs), and exogenous AIBP was much less effective in T cells than in MDMs. AIBP inhibited virus-cell fusion, specifically targeting cells with lipid rafts mobilized by cell activation or Nef-containing exosomes. MDM-HIV fusion was sensitive to AIBP only in the presence of Nef provided by the virus or exosomes. Peripheral blood mononuclear cells from donors with the HLA-B*35 genotype, associated with rapid progression of HIV disease, bound less AIBP than cells from donors with other HLA genotypes and were not protected by AIBP from rapid HIV-1 replication. These results provide the first evidence for the role of Nef exosomes in regulating HIV-cell fusion by modifying lipid rafts and suggest that AIBP is an innate factor that restricts HIV replication by targeting lipid rafts.
Collapse
|
10
|
IL-2 receptors preassemble and signal in the ER/Golgi causing resistance to antiproliferative anti-IL-2Rα therapies. Proc Natl Acad Sci U S A 2019; 116:21120-21130. [PMID: 31570576 DOI: 10.1073/pnas.1901382116] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Interleukin-2 (IL-2) and IL-15 play pivotal roles in T cell activation, apoptosis, and survival, and are implicated in leukemias and autoimmune diseases. Their heterotrimeric receptors share their β- and γc-chains, but have distinct α-chains. Anti-IL-2Rα (daclizumab) therapy targeting cell surface-expressed receptor subunits to inhibit T cell proliferation has only brought limited success in adult T cell leukemia/lymphoma (ATL) and in multiple sclerosis. We asked whether IL-2R subunits could already preassemble and signal efficiently in the endoplasmic reticulum (ER) and the Golgi. A combination of daclizumab and anti-IL-2 efficiently blocked IL-2-induced proliferation of IL-2-dependent wild-type (WT) ATL cells but not cells transfected with IL-2, suggesting that in IL-2-producing cells signaling may already take place before receptors reach the cell surface. In the Golgi fraction isolated from IL-2-producing ATL cells, we detected by Western blot phosphorylated Jak1, Jak3, and a phosphotyrosine signal attributed to the γc-chain, which occurred at much lower levels in the Golgi of WT ATL cells. We expressed EGFP- and mCherry-tagged receptor chains in HeLa cells to study their assembly along the secretory pathway. Confocal microscopy, Förster resonance energy transfer, and imaging fluorescence cross-correlation spectroscopy analysis revealed partial colocalization and molecular association of IL-2 (and IL-15) receptor chains in the ER/Golgi, which became more complete in the plasma membrane, further confirming our hypothesis. Our results define a paradigm of intracellular autocrine signaling and may explain resistance to antagonistic antibody therapies targeting receptors at the cell surface.
Collapse
|
11
|
Dominant role of splenic marginal zone lipid rafts in the classical complement pathway against S. pneumoniae. Cell Death Discov 2019; 5:133. [PMID: 31531231 PMCID: PMC6733876 DOI: 10.1038/s41420-019-0213-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/11/2019] [Accepted: 08/18/2019] [Indexed: 12/28/2022] Open
Abstract
Lipid rafts (LRs) play crucial roles in complex physiological processes, modulating innate and acquired immune responses to pathogens. The transmembrane C-type lectins human dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN) and its mouse homolog SIGN-R1 are distributed in LRs and expressed on splenic marginal zone (MZ) macrophages. The DC-SIGN-C1q or SIGN-R1-C1q complex could mediate the immunoglobulin (Ig)-independent classical complement pathway against Streptococcus pneumoniae. Precise roles of LRs during this complement pathway are unknown. Here we show that LRs are indispensable for accelerating the DC-SIGN- or SIGN-R1-mediated classical complement pathway against S. pneumoniae, thus facilitating rapid clearance of the pathogen. The trimolecular complex of SIGN-R1-C1q-C4 was exclusively enriched in LRs of splenic MZ macrophages and their localization was essential for activating C3 catabolism and enhancing pneumococcal clearance, which were abolished in SIGN-R1-knockout mice. However, DC-SIGN replacement on splenic MZ macrophage’s LRs of SIGN-R1-depleted mice reversed these defects. Disruption of LRs dramatically reduced pneumococcal uptake and decomposition. Additionally, DC- SIGN, C1q, C4, and C3 were obviously distributed in splenic LRs of cadavers. Therefore, LRs on splenic SIGN-R1+ or DC-SIGN+ macrophages could provide spatially confined and optimal bidirectional platforms, not only for usual intracellular events, for example recognition and phagocytosis of pathogens, but also an unusual extracellular event such as the complement system. These findings improve our understanding of the orchestrated roles of the spleen, unraveling a new innate immune system initiated from splenic MZ LRs, and yielding answers to several long-standing problems, including the need to understand the profound role of LRs in innate immunity, the need to identify how such a small portion of splenic SIGN-R1+ macrophages (<0.05% of splenic macrophages) effectively resist S. pneumoniae, and the need to understand how LRs can promote the protective function of DC-SIGN against S. pneumoniae in the human spleen.
Collapse
|
12
|
Grosjean K, Der C, Robert F, Thomas D, Mongrand S, Simon-Plas F, Gerbeau-Pissot P. Interactions between lipids and proteins are critical for organization of plasma membrane-ordered domains in tobacco BY-2 cells. JOURNAL OF EXPERIMENTAL BOTANY 2018; 69:3545-3557. [PMID: 29722895 PMCID: PMC6022670 DOI: 10.1093/jxb/ery152] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 04/16/2018] [Indexed: 05/20/2023]
Abstract
The laterally heterogeneous plant plasma membrane (PM) is organized into finely controlled specialized areas that include membrane-ordered domains. Recently, the spatial distribution of such domains within the PM has been identified as playing a key role in cell responses to environmental challenges. To examine membrane order at a local level, BY-2 tobacco suspension cell PMs were labelled with an environment-sensitive probe (di-4-ANEPPDHQ). Four experimental models were compared to identify mechanisms and cell components involved in short-term (1 h) maintenance of the ordered domain organization in steady-state cell PMs: modulation of the cytoskeleton or the cell wall integrity of tobacco BY-2 cells; and formation of giant vesicles using either a lipid mixture of tobacco BY-2 cell PMs or the original lipid and protein combinations of the tobacco BY-2 cell PM. Whilst inhibiting phosphorylation or disrupting either the cytoskeleton or the cell wall had no observable effects, we found that lipids and proteins significantly modified both the abundance and spatial distribution of ordered domains. This indicates the involvement of intrinsic membrane components in the local physical state of the plant PM. Our findings support a major role for the 'lipid raft' model, defined as the sterol-dependent ordered assemblies of specific lipids and proteins in plant PM organization.
Collapse
Affiliation(s)
- Kevin Grosjean
- Agroécologie, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | - Christophe Der
- Agroécologie, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | - Franck Robert
- Agroécologie, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | - Dominique Thomas
- Agroécologie, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | - Sébastien Mongrand
- Laboratoire de Biogenèse Membranaire (LBM), Unité Mixte de Recherche UMR, CNRS, Université de Bordeaux, Bordeaux, France
| | - Françoise Simon-Plas
- Agroécologie, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | | |
Collapse
|
13
|
Nagy É, Mocsár G, Sebestyén V, Volkó J, Papp F, Tóth K, Damjanovich S, Panyi G, Waldmann TA, Bodnár A, Vámosi G. Membrane Potential Distinctly Modulates Mobility and Signaling of IL-2 and IL-15 Receptors in T Cells. Biophys J 2018; 114:2473-2482. [PMID: 29754714 PMCID: PMC6129476 DOI: 10.1016/j.bpj.2018.04.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/21/2018] [Accepted: 04/23/2018] [Indexed: 12/11/2022] Open
Abstract
The high electric field across the plasma membrane might influence the conformation and behavior of transmembrane proteins that have uneven charge distributions in or near their transmembrane regions. Membrane depolarization of T cells occurs in the tumor microenvironment and in inflamed tissues because of K+ release from necrotic cells and hypoxia affecting the expression of K+ channels. However, little attention has been given to the effect of membrane potential (MP) changes on membrane receptor function. Therefore, we studied the influence of membrane de- and hyperpolarization on the biophysical properties and signaling of interleukin-2 (IL-2) and interleukin-15 (IL-15) receptors, which play important roles in T cell function. We investigated the mobility, clustering, and signaling of these receptors and major histocompatibility complex (MHC) I/II glycoproteins forming coclusters in lipid rafts of T cells. Depolarization by high K+ buffer or K+ channel blockers resulted in a decrease in the mobility of IL-2Rα and MHC glycoproteins, as shown by fluorescence correlation spectroscopy, whereas hyperpolarization by the K+ ionophore valinomycin increased their mobility. Contrary to this, the mobility of IL-15Rα decreased upon both de- and hyperpolarization. These changes in protein mobility are not due to an alteration of membrane fluidity, as evidenced by fluorescence anisotropy measurements. Förster resonance energy transfer measurements showed that most homo- or heteroassociations of IL-2R, IL-15R, and MHC I did not change considerably, either. MP changes modulated signaling by the two cytokines in distinct ways: depolarization caused a significant increase in the IL-2-induced phosphorylation of signal transducer and activator of transcription 5, whereas hyperpolarization evoked a decrease only in the IL-15-induced signal. Our data imply that the MP may be an important modulator of interleukin receptor signaling and dynamics. Enhanced IL-2 signaling in depolarized Treg cells highly expressing IL-2R may contribute to suppression of antitumor immune surveillance.
Collapse
Affiliation(s)
- Éva Nagy
- Department of Biophysics and Cell Biology, Faculty of Medicine
| | - Gábor Mocsár
- Department of Biophysics and Cell Biology, Faculty of Medicine
| | | | - Julianna Volkó
- Department of Biophysics and Cell Biology, Faculty of Medicine
| | - Ferenc Papp
- Department of Biophysics and Cell Biology, Faculty of Medicine; MTA-DE- NAP B Ion Channel Structure-Function Research Group, RCMM, University of Debrecen, Debrecen, Hungary
| | - Katalin Tóth
- Division Biophysics of Macromolecules, German Cancer Research Center, Heidelberg, Germany
| | | | - György Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine; MTA-DE- NAP B Ion Channel Structure-Function Research Group, RCMM, University of Debrecen, Debrecen, Hungary
| | - Thomas A Waldmann
- Lymphoid Malignancies Branch, National Institutes of Health, Bethesda, Maryland
| | - Andrea Bodnár
- Department of Biophysics and Cell Biology, Faculty of Medicine
| | - György Vámosi
- Department of Biophysics and Cell Biology, Faculty of Medicine.
| |
Collapse
|
14
|
Lu Y, Tan CTY, Nyunt MSZ, Mok EWH, Camous X, Kared H, Fulop T, Feng L, Ng TP, Larbi A. Inflammatory and immune markers associated with physical frailty syndrome: findings from Singapore longitudinal aging studies. Oncotarget 2018; 7:28783-95. [PMID: 27119508 PMCID: PMC5045356 DOI: 10.18632/oncotarget.8939] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 04/10/2016] [Indexed: 12/31/2022] Open
Abstract
Chronic systematic inflammation and reduced immune system fitness are considered potential contributing factors to the development of age-related frailty, but the underlying mechanisms are poorly defined. This exploratory study aimed to identify frailty-related inflammatory markers and immunological phenotypes in a cohort of community-dwelling adults aged ≥ 55 years. Frailty was assessed using two models, a Frailty Index and a categorical phenotype, and correlated with levels of circulating immune biomarkers and markers of senescence in immune cell subsets. We identified eight serological biomarkers that were associated with frailty, including sgp130, IL-2Rα, I-309, MCP-1, BCA-1, RANTES, leptin, and IL-6R. Frailty Index was inversely predicted by the frequency of CD3+, CD45RA+, and central memory CD4 cells, and positively predicted by the loss of CD28 expression, especially in CD8+ T cells, while frailty status was predicted by the frequency of terminal effector CD8+ T cells. In γ/δ T cells, frailty was negatively associated with CD27, and positively associated with IFNγ+TNFα- secretion by γ/δ2+ cells and IFNγ-TNFα+ secretion by γ/δ2- cells. Increased numbers of exhausted and CD38+ B cells, as well as CD14+CD16+ inflammatory monocytes, were also identified as frailty-associated phenotypes. This pilot study supports an association between inflammation, cellular immunity, and the process of frailty. These findings have significance for the early identification of frailty using circulating biomarkers prior to clinical manifestations of severe functional decline in the elderly.
Collapse
Affiliation(s)
- Yanxia Lu
- Department of Clinical Psychology and Psychiatry/School of Public Health, Zhejiang University College of Medicine, Hangzhou, China
| | - Crystal Tze Ying Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Ma Shwe Zin Nyunt
- Gerontology Research Programme, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Esther Wing Hei Mok
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Xavier Camous
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Hassen Kared
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Tamas Fulop
- Geriatrics Division, Department of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Liang Feng
- Graduate Medical School, Duke-National University of Singapore, Singapore
| | - Tze Pin Ng
- Gerontology Research Programme, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore.,Geriatrics Division, Department of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
15
|
Bitler A, Dover RS, Shai Y. Fractal properties of cell surface structures: A view from AFM. Semin Cell Dev Biol 2018; 73:64-70. [DOI: 10.1016/j.semcdb.2017.07.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/10/2017] [Accepted: 07/13/2017] [Indexed: 01/08/2023]
|
16
|
Mocsár G, Volkó J, Rönnlund D, Widengren J, Nagy P, Szöllősi J, Tóth K, Goldman CK, Damjanovich S, Waldmann TA, Bodnár A, Vámosi G. MHC I Expression Regulates Co-clustering and Mobility of Interleukin-2 and -15 Receptors in T Cells. Biophys J 2017; 111:100-12. [PMID: 27410738 DOI: 10.1016/j.bpj.2016.05.044] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 05/09/2016] [Accepted: 05/16/2016] [Indexed: 11/28/2022] Open
Abstract
MHC glycoproteins form supramolecular clusters with interleukin-2 and -15 receptors in lipid rafts of T cells. The role of highly expressed MHC I in maintaining these clusters is unknown. We knocked down MHC I in FT7.10 human T cells, and studied protein clustering at two hierarchic levels: molecular aggregations and mobility by Förster resonance energy transfer and fluorescence correlation spectroscopy; and segregation into larger domains or superclusters by superresolution stimulated emission depletion microscopy. Fluorescence correlation spectroscopy-based molecular brightness analysis revealed that the studied molecules diffused as tight aggregates of several proteins of a kind. Knockdown reduced the number of MHC I containing molecular aggregates and their average MHC I content, and decreased the heteroassociation of MHC I with IL-2Rα/IL-15Rα. The mobility of not only MHC I but also that of IL-2Rα/IL-15Rα increased, corroborating the general size decrease of tight aggregates. A multifaceted analysis of stimulated emission depletion images revealed that the diameter of MHC I superclusters diminished from 400-600 to 200-300 nm, whereas those of IL-2Rα/IL-15Rα hardly changed. MHC I and IL-2Rα/IL-15Rα colocalized with GM1 ganglioside-rich lipid rafts, but MHC I clusters retracted to smaller subsets of GM1- and IL-2Rα/IL-15Rα-rich areas upon knockdown. Our results prove that changes in expression level may significantly alter the organization and mobility of interacting membrane proteins.
Collapse
Affiliation(s)
- Gábor Mocsár
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Julianna Volkó
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Daniel Rönnlund
- Department of Applied Physics/Experimental Biomolecular Physics, Royal Institute of Technology, Albanova University Center, Stockholm, Sweden
| | - Jerker Widengren
- Department of Applied Physics/Experimental Biomolecular Physics, Royal Institute of Technology, Albanova University Center, Stockholm, Sweden
| | - Péter Nagy
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Szöllősi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Cell Biology and Signaling Research Group of the Hungarian Academy of Sciences and the University of Debrecen, Debrecen, Hungary
| | - Katalin Tóth
- German Cancer Research Center, Biophysics of Macromolecules, Heidelberg, Germany
| | - Carolyn K Goldman
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sándor Damjanovich
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Thomas A Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Andrea Bodnár
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Vámosi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
17
|
Dengue Virus Infection Is through a Cooperative Interaction between a Mannose Receptor and CLEC5A on Macrophage as a Multivalent Hetero-Complex. PLoS One 2016; 11:e0166474. [PMID: 27832191 PMCID: PMC5104462 DOI: 10.1371/journal.pone.0166474] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 10/28/2016] [Indexed: 01/01/2023] Open
Abstract
Dengue fever is a mosquito-borne viral pandemic disease that is widespread in the tropical and subtropical areas. Dengue virus uses human mannose-binding receptor (MR) and DC-SIGN on macrophages as primary receptors, and CLEC5A as signaling receptor to sense the dengue virus invasion and then to signal and stimulate macrophages to secrete cytokines. But the interplay between MR/DC-SIGN and CLEC5A is unknown. Here we demonstrate a plausible mechanism for the interaction, i.e. MR/DC-SIGN first attracts the virus with high avidity, and the virus concurrently interacts with CLEC5A in close proximity to form a multivalent hetero-complex and facilitate CLEC5A-mediated signal transduction. Our study suggests that the cooperation between a high-avidity lectin-virus interaction and a nearby low-avidity signaling receptor provides a necessary connection between binding and signaling. Understanding this mechanism may lead to the development of a new antiviral strategy.
Collapse
|
18
|
Mattila PK, Batista FD, Treanor B. Dynamics of the actin cytoskeleton mediates receptor cross talk: An emerging concept in tuning receptor signaling. J Cell Biol 2016; 212:267-80. [PMID: 26833785 PMCID: PMC4748574 DOI: 10.1083/jcb.201504137] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Recent evidence implicates the actin cytoskeleton in the control of receptor signaling. This may be of particular importance in the context of immune receptors, such as the B cell receptor, where dysregulated signaling can result in autoimmunity and malignancy. Here, we discuss the role of the actin cytoskeleton in controlling receptor compartmentalization, dynamics, and clustering as a means to regulate receptor signaling through controlling the interactions with protein partners. We propose that the actin cytoskeleton is a point of integration for receptor cross talk through modulation of protein dynamics and clustering. We discuss the implication of this cross talk via the cytoskeleton for both ligand-induced and low-level constitutive (tonic) signaling necessary for immune cell survival.
Collapse
Affiliation(s)
- Pieta K Mattila
- Institute of Biomedicine, MediCity, University of Turku, 20520 Turku, Finland
| | - Facundo D Batista
- Lymphocyte Interaction Laboratory, The Francis Crick Institute, Cancer Research UK, London WC2A 3LY, England, UK
| | - Bebhinn Treanor
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario M5T 1C6, Canada
| |
Collapse
|
19
|
Annexin A6 regulates interleukin-2-mediated T-cell proliferation. Immunol Cell Biol 2016; 94:543-53. [PMID: 26853809 DOI: 10.1038/icb.2016.15] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 12/10/2015] [Accepted: 01/10/2016] [Indexed: 02/06/2023]
Abstract
Annexin A6 (AnxA6) has been implicated in cell signalling by contributing to the organisation of the plasma membrane. Here we examined whether AnxA6 regulates signalling and proliferation in T cells. We used a contact hypersensitivity model to immune challenge wild-type (WT) and AnxA6(-/-) mice and found that the in vivo proliferation of CD4(+) T cells, but not CD8(+) T cells, was impaired in AnxA6(-/-) relative to WT mice. However, T-cell migration and signalling through the T-cell receptor ex vivo was similar between T cells isolated from AnxA6(-/-) and WT mice. In contrast, interleukin-2 (IL-2) signalling was reduced in AnxA6(-/-) compared with WT T cells. Further, AnxA6-deficient T cells had reduced membrane order and cholesterol levels. Taken together, our data suggest that AnxA6 regulates IL-2 homeostasis and sensitivity in T cells by sustaining a lipid raft-like membrane environment.
Collapse
|
20
|
Szöőr Á, Ujlaky-Nagy L, Tóth G, Szöllősi J, Vereb G. Cell confluence induces switching from proliferation to migratory signaling by site-selective phosphorylation of PDGF receptors on lipid raft platforms. Cell Signal 2015; 28:81-93. [PMID: 26631574 DOI: 10.1016/j.cellsig.2015.11.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 11/25/2015] [Accepted: 11/25/2015] [Indexed: 10/22/2022]
Abstract
Platelet derived growth factor receptors (PDGFR) play an important role in tumor pathogenesis and are frequently overexpressed in glioblastoma. Earlier we have shown that only confluent glioblastoma cell cultures exhibit a biphasic calcium transient upon PDGF stimulation. Here, we examined how the change in cell density leads to differential cellular responses to the same PDGF stimulus. PDGF beta receptors and their specific phosphotyrosine residues were fluorescently co-labeled on A172 and T98G glioblastoma cells. The distribution in cell membrane microdomains (lipid rafts) and the phosphorylation state of PDGFR was measured by confocal microscopy and quantitated by digital image processing. Corresponding bulk data were obtained by Western blotting. Activation of relevant downstream signaling pathways was assessed by immunofluorescence in confocal microscopy and by Western blot analysis. Functional outcomes were confirmed with bulk and single cell proliferation assays and motility measurements. In non-confluent (sparse) cultures PDGF-BB stimulation significantly increased phosphorylation of Tyr716 specific for the Ras/MAPK pathway and Tyr751 specific for the phosphoinositide 3-kinase/Akt pathway. As cell monolayers reached confluence, Tyr771 and Tyr1021 were the prominently phosphorylated residues. Tyr771 serves as adaptor for Ras-GAP, which inactivates the MAPK pathway, and Tyr1021 feeds into the phospholipase C-gamma/PKC pathway. Coherent with this, MAPK phosphorylation, Ki-67 positivity and proliferation dominated in dispersed cells, and could be abolished with inhibitors of the MAPK pathway. At the same time, RhoA activation, redistribution of cortactin to leading edges, and increased motility were the prominent output features in confluent cultures. Importantly, the stimulus-evoked confluence-specific changes in the phosphorylation of tyrosine residues occurred mainly in GM1-rich lipid microdomains (rafts). These observations suggest that the same stimulus is able to promote distinctly relevant signaling outputs through a confluence dependent, lipid raft-based regulatory mechanism. In particular, cell division and survival in sparse cultures and inhibition of proliferation and promotion of migration in confluent monolayers. In our model, the ability to switch the final output of the same stimulus as a function of cell density could be a key to the balance of proliferation and invasion in malignant glioblastoma.
Collapse
Affiliation(s)
- Árpád Szöőr
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Ujlaky-Nagy
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Tóth
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Szöllősi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Vereb
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
21
|
Gao J, Wang Y, Cai M, Pan Y, Xu H, Jiang J, Ji H, Wang H. Mechanistic insights into EGFR membrane clustering revealed by super-resolution imaging. NANOSCALE 2015; 7:2511-9. [PMID: 25569174 DOI: 10.1039/c4nr04962d] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The clustering of membrane receptors such as EGFR is critical for various biological processes, for example cell signaling and tumorigenesis. However, the mechanism involved remains poorly understood. Here, we used a super resolution imaging technique, which has shattered the longstanding resolution barrier of light diffraction, to investigate the distribution of membrane EGFR on apical or basal surfaces of COS-7 cells and on the surface of suspended COS-7 cells. Our data show that more and larger EGFR clusters are detected on the apical surface in comparison with those on the basal surface and this difference is not affected by the EGFR activation state, whereas suspended COS-7 cells exhibit a moderate clustering state and a homogeneous distribution pattern, indicating that the external environment surrounding the cell membrane is the decisive factor in the EGFR clustering pattern. A dual-color dSTORM image reveals the significant colocalization of EGFR and lipid rafts; interestingly MβCD treatment leads to a dramatic decrease of the amount and size of EGFR clusters on both apical and basal surfaces, highlighting a key role of lipid rafts in EGFR cluster formation. Altogether, our results illustrate the distribution pattern of EGFR in polarized cells and uncover the essential role of lipid rafts in EGFR cluster maintenance.
Collapse
Affiliation(s)
- Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P.R. China.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Nizsalóczki E, Csomós I, Nagy P, Fazekas Z, Goldman CK, Waldmann TA, Damjanovich S, Vámosi G, Mátyus L, Bodnár A. Distinct spatial relationship of the interleukin-9 receptor with interleukin-2 receptor and major histocompatibility complex glycoproteins in human T lymphoma cells. Chemphyschem 2014; 15:3969-78. [PMID: 25297818 DOI: 10.1002/cphc.201402501] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 08/16/2014] [Indexed: 11/07/2022]
Abstract
The interleukin-9 receptor (IL-9R) consists of an α subunit and a γ(c) chain that are shared with other cytokine receptors, including interleukin-2 receptor (IL-2R), an important regulator of T cells. We previously showed that IL-2R is expressed in common clusters with major histocompatibility complex (MHC) glycoproteins in lipid rafts of human T lymphoma cells, which raised the question about what the relationship between clusters of IL-2R/MHC and IL-9R is. Confocal microscopy colocalization and fluorescence resonance energy transfer experiments capable of detecting membrane protein organization at different size scales revealed nonrandom association of IL-9R with IL-2R/MHC clusters at the surface of human T lymphoma cells. Accommodation of IL-9Rα in membrane areas segregated from the IL-2R/MHC domains was also detected. The bipartite nature of IL-9R distribution was mirrored by signal transducer and activator of transcription (STAT) activation results. Our data indicate that co-compartmentalization with MHC glycoproteins is a general property of γ(c) receptors. Distribution of receptor chains between different membrane domains may regulate their function.
Collapse
Affiliation(s)
- Enikő Nizsalóczki
- Department of Biophysics and Cell Biology, Research Center for Molecular Medicine, University of Debrecen, P.O.B. 39., 4012, Debrecen (Hungary)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Anderson HA, Roche PA. MHC class II association with lipid rafts on the antigen presenting cell surface. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:775-80. [PMID: 25261705 DOI: 10.1016/j.bbamcr.2014.09.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 09/16/2014] [Accepted: 09/17/2014] [Indexed: 12/29/2022]
Abstract
MHC class II (MHC-II) molecules function by binding peptides derived from either self or foreign proteins and expressing these peptides on the surface of antigen presenting cells (APCs) for recognition by CD4 T cells. MHC-II is known to exist on clusters on the surface of APCs, and a variety of biochemical and functional studies have suggested that these clusters represent lipid raft microdomain-associated MHC-II. This review will summarize data exploring the biosynthesis of raft-associated MHC-II and the role that lipid raft association plays in regulating T cell activation by APCs. This article is part of a Special Issue entitled: Nanoscale membrane organisation and signalling.
Collapse
Affiliation(s)
- Howard A Anderson
- Division of Therapeutic Proteins, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Paul A Roche
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
24
|
Single-laser polarization FRET (polFRET) on the cell surface. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:3047-64. [PMID: 25241341 DOI: 10.1016/j.bbamcr.2014.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 07/15/2014] [Accepted: 07/21/2014] [Indexed: 11/22/2022]
Abstract
A new method for the simultaneous detection of rotational mobility and proximity of cell surface receptors is presented based on cell-by-cell basis measurement of polarized fluorescence intensity components of the donor and acceptor of a FRET system. In addition to the FRET efficiency and the donor and acceptor concentrations, the method makes also possible the determination of the rotational characteristics and the associated fraction of the donors (FRET-fraction). The method is illustrated with flow cytometric and rFLIM measurements on donor-acceptor systems comprising fluorescently labeled whole antibodies and their Fab fragments against epitopes of the MHCI and MHCII cell surface receptors on human lymphoblast cells. Fluorescence anisotropy of donor and acceptor and FRET efficiency were measured for samples of different acceptor-to-donor concentration ratios. Acceptor anisotropy proved to be more sensitive than the donor anisotropy for sensing FRET. After determining the rotational constants of the donor-conjugated antibodies by measurements of FRET in the steady state, and by rFLIM as a reference, the associated fractions of the MHCI and MHCII molecules in their clusters were determined. Besides the flow cytometer and the wide-field rFLIM used in this study, the method can be applied also in other devices capable of dual-anisotropy detection.
Collapse
|
25
|
Mocanu MM, Ganea C, Georgescu L, Váradi T, Shrestha D, Baran I, Katona E, Nagy P, Szöllősi J. Epigallocatechin 3-O-gallate induces 67 kDa laminin receptor-mediated cell death accompanied by downregulation of ErbB proteins and altered lipid raft clustering in mammary and epidermoid carcinoma cells. JOURNAL OF NATURAL PRODUCTS 2014; 77:250-7. [PMID: 24456004 DOI: 10.1021/np4007712] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Since the administration of synthetic medicines is associated with drug resistance and undesired side effects, utilization of natural compounds could be an alternative and complementary modality to inhibit or prevent the development of tumors. Epigallocatechin 3-O-gallate (EGCG, 1), the major flavan component of green tea, and genistein (2), a soy isoflavonoid, are known to have chemopreventive and chemotherapeutic effects against cancer. This study demonstrated that both flavonoids inhibit cell proliferation, an effect enhanced under serum-free conditions. Compound 1, but not 2, induced downregulation of ErbB1 and ErbB2 in mammary and epidermoid carcinoma cells, and its inhibitory effect on cell viability was mediated by the 67 kDa laminin receptor (67LR). While 1 was superior in inducing cell death, 2 was more efficient in arresting the tumor cells in the G2/M phase. Furthermore, number and brightness analysis revealed that 1 decreased the homoclustering of a lipid raft marker, glycosylphosphatidylinositol-anchored GFP, and it also reduced the co-localization between lipid rafts and 67LR. The main conclusion made is that the primary target of 1 may be the lipid raft component of the plasma membrane followed by secondary changes in the expression of ErbB proteins. Compound 2, on the other hand, must have other unidentified targets.
Collapse
Affiliation(s)
- Maria-Magdalena Mocanu
- Department of Biophysics, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy , 050474 Bucharest, Romania
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Reuven EM, Fink A, Shai Y. Regulation of innate immune responses by transmembrane interactions: lessons from the TLR family. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:1586-93. [PMID: 24480409 DOI: 10.1016/j.bbamem.2014.01.020] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 01/15/2014] [Accepted: 01/18/2014] [Indexed: 02/06/2023]
Abstract
The mammalian innate immune response is responsible for the early stages of defense against invading pathogens. One of the major receptor families facilitating innate immune activation is the Toll-like receptor (TLR) family. These receptors are type 1 membrane proteins spanning the membrane with a single transmembrane domain (TMD). All TLRs form homo- and hetero-dimers within membranes and new data suggest that the single transmembrane domain of some of these receptors is involved in their dimerization and function. Newly identified TLR dimers are continuously reported but only little is known about the importance of the TMDs for their dimer assembly and signaling regulation. Uncontrolled or untimely activation of TLRs is related to a large number of pathologies ranging from cystic fibrosis to sepsis and cancer. In this review we will focus on the contribution of the TMDs of innate immune receptors - specifically TLR2-to their regulation and function. In addition, we will address the current issues remaining to be solved regarding the mechanistic insights of this regulation. This article is part of a Special Issue entitled: Membrane Structure and Function: Relevance in the Cell's Physiology, Pathology and Therapy.
Collapse
Affiliation(s)
- Eliran Moshe Reuven
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Avner Fink
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yechiel Shai
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
27
|
CD1d favors MHC neighborhood, GM1 ganglioside proximity and low detergent sensitive membrane regions on the surface of B lymphocytes. Biochim Biophys Acta Gen Subj 2014. [DOI: 10.1016/j.bbagen.2013.10.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
28
|
Gerbeau-Pissot P, Der C, Thomas D, Anca IA, Grosjean K, Roche Y, Perrier-Cornet JM, Mongrand S, Simon-Plas F. Modification of plasma membrane organization in tobacco cells elicited by cryptogein. PLANT PHYSIOLOGY 2014; 164:273-86. [PMID: 24235133 PMCID: PMC3875808 DOI: 10.1104/pp.113.225755] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 11/05/2013] [Indexed: 05/07/2023]
Abstract
Lipid mixtures within artificial membranes undergo a separation into liquid-disordered and liquid-ordered phases. However, the existence of this segregation into microscopic liquid-ordered phases has been difficult to prove in living cells, and the precise organization of the plasma membrane into such phases has not been elucidated in plant cells. We developed a multispectral confocal microscopy approach to generate ratiometric images of the plasma membrane surface of Bright Yellow 2 tobacco (Nicotiana tabacum) suspension cells labeled with an environment sensitive fluorescent probe. This allowed the in vivo characterization of the global level of order of this membrane, by which we could demonstrate that an increase in its proportion of ordered phases transiently occurred in the early steps of the signaling triggered by cryptogein and flagellin, two elicitors of plant defense reactions. The use of fluorescence recovery after photobleaching revealed an increase in plasma membrane fluidity induced by cryptogein, but not by flagellin. Moreover, we characterized the spatial distribution of liquid-ordered phases on the membrane of living plant cells and monitored their variations induced by cryptogein elicitation. We analyze these results in the context of plant defense signaling, discuss their meaning within the framework of the "membrane raft" hypothesis, and propose a new mechanism of signaling platform formation in response to elicitor treatment.
Collapse
Affiliation(s)
| | - Christophe Der
- Université de Bourgogne (P.G.-P., C.D., D.T., K.G.), and Institut National de la Recherche Agronomique (I.-A.A., Y.R., F.S.-P.), Unité Mixte de Recherche 1347 Agroécologie, Equipe de Recherche Labelisée 6300 Centre National de la Recherche Scientifique, BP 86510, F–21000 Dijon, France
- AgroSup Dijon, Laboratoire Procédés Alimentaires et Microbiologiques, F–21000 Dijon, France (J.-M.P.-C.); and
- Centre National de la Recherche Scientifique, Laboratoire de Biogenèse Membranaire, Unité Mixte de Recherche 5200, Université Victor Segalen, Institut National de la Recherche Agronomique Bordeaux Aquitaine, BP 81, F–33883 Villenave d’Ornon, France (S.M.)
| | - Dominique Thomas
- Université de Bourgogne (P.G.-P., C.D., D.T., K.G.), and Institut National de la Recherche Agronomique (I.-A.A., Y.R., F.S.-P.), Unité Mixte de Recherche 1347 Agroécologie, Equipe de Recherche Labelisée 6300 Centre National de la Recherche Scientifique, BP 86510, F–21000 Dijon, France
- AgroSup Dijon, Laboratoire Procédés Alimentaires et Microbiologiques, F–21000 Dijon, France (J.-M.P.-C.); and
- Centre National de la Recherche Scientifique, Laboratoire de Biogenèse Membranaire, Unité Mixte de Recherche 5200, Université Victor Segalen, Institut National de la Recherche Agronomique Bordeaux Aquitaine, BP 81, F–33883 Villenave d’Ornon, France (S.M.)
| | - Iulia-Andra Anca
- Université de Bourgogne (P.G.-P., C.D., D.T., K.G.), and Institut National de la Recherche Agronomique (I.-A.A., Y.R., F.S.-P.), Unité Mixte de Recherche 1347 Agroécologie, Equipe de Recherche Labelisée 6300 Centre National de la Recherche Scientifique, BP 86510, F–21000 Dijon, France
- AgroSup Dijon, Laboratoire Procédés Alimentaires et Microbiologiques, F–21000 Dijon, France (J.-M.P.-C.); and
- Centre National de la Recherche Scientifique, Laboratoire de Biogenèse Membranaire, Unité Mixte de Recherche 5200, Université Victor Segalen, Institut National de la Recherche Agronomique Bordeaux Aquitaine, BP 81, F–33883 Villenave d’Ornon, France (S.M.)
| | - Kevin Grosjean
- Université de Bourgogne (P.G.-P., C.D., D.T., K.G.), and Institut National de la Recherche Agronomique (I.-A.A., Y.R., F.S.-P.), Unité Mixte de Recherche 1347 Agroécologie, Equipe de Recherche Labelisée 6300 Centre National de la Recherche Scientifique, BP 86510, F–21000 Dijon, France
- AgroSup Dijon, Laboratoire Procédés Alimentaires et Microbiologiques, F–21000 Dijon, France (J.-M.P.-C.); and
- Centre National de la Recherche Scientifique, Laboratoire de Biogenèse Membranaire, Unité Mixte de Recherche 5200, Université Victor Segalen, Institut National de la Recherche Agronomique Bordeaux Aquitaine, BP 81, F–33883 Villenave d’Ornon, France (S.M.)
| | - Yann Roche
- Université de Bourgogne (P.G.-P., C.D., D.T., K.G.), and Institut National de la Recherche Agronomique (I.-A.A., Y.R., F.S.-P.), Unité Mixte de Recherche 1347 Agroécologie, Equipe de Recherche Labelisée 6300 Centre National de la Recherche Scientifique, BP 86510, F–21000 Dijon, France
- AgroSup Dijon, Laboratoire Procédés Alimentaires et Microbiologiques, F–21000 Dijon, France (J.-M.P.-C.); and
- Centre National de la Recherche Scientifique, Laboratoire de Biogenèse Membranaire, Unité Mixte de Recherche 5200, Université Victor Segalen, Institut National de la Recherche Agronomique Bordeaux Aquitaine, BP 81, F–33883 Villenave d’Ornon, France (S.M.)
| | - Jean-Marie Perrier-Cornet
- Université de Bourgogne (P.G.-P., C.D., D.T., K.G.), and Institut National de la Recherche Agronomique (I.-A.A., Y.R., F.S.-P.), Unité Mixte de Recherche 1347 Agroécologie, Equipe de Recherche Labelisée 6300 Centre National de la Recherche Scientifique, BP 86510, F–21000 Dijon, France
- AgroSup Dijon, Laboratoire Procédés Alimentaires et Microbiologiques, F–21000 Dijon, France (J.-M.P.-C.); and
- Centre National de la Recherche Scientifique, Laboratoire de Biogenèse Membranaire, Unité Mixte de Recherche 5200, Université Victor Segalen, Institut National de la Recherche Agronomique Bordeaux Aquitaine, BP 81, F–33883 Villenave d’Ornon, France (S.M.)
| | - Sébastien Mongrand
- Université de Bourgogne (P.G.-P., C.D., D.T., K.G.), and Institut National de la Recherche Agronomique (I.-A.A., Y.R., F.S.-P.), Unité Mixte de Recherche 1347 Agroécologie, Equipe de Recherche Labelisée 6300 Centre National de la Recherche Scientifique, BP 86510, F–21000 Dijon, France
- AgroSup Dijon, Laboratoire Procédés Alimentaires et Microbiologiques, F–21000 Dijon, France (J.-M.P.-C.); and
- Centre National de la Recherche Scientifique, Laboratoire de Biogenèse Membranaire, Unité Mixte de Recherche 5200, Université Victor Segalen, Institut National de la Recherche Agronomique Bordeaux Aquitaine, BP 81, F–33883 Villenave d’Ornon, France (S.M.)
| | - Françoise Simon-Plas
- Université de Bourgogne (P.G.-P., C.D., D.T., K.G.), and Institut National de la Recherche Agronomique (I.-A.A., Y.R., F.S.-P.), Unité Mixte de Recherche 1347 Agroécologie, Equipe de Recherche Labelisée 6300 Centre National de la Recherche Scientifique, BP 86510, F–21000 Dijon, France
- AgroSup Dijon, Laboratoire Procédés Alimentaires et Microbiologiques, F–21000 Dijon, France (J.-M.P.-C.); and
- Centre National de la Recherche Scientifique, Laboratoire de Biogenèse Membranaire, Unité Mixte de Recherche 5200, Université Victor Segalen, Institut National de la Recherche Agronomique Bordeaux Aquitaine, BP 81, F–33883 Villenave d’Ornon, France (S.M.)
| |
Collapse
|
29
|
Identification of cell-surface molecular interactions under living conditions by using the enzyme-mediated activation of radical sources (EMARS) method. SENSORS 2012; 12:16037-45. [PMID: 23443365 PMCID: PMC3571769 DOI: 10.3390/s121216037] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 11/12/2012] [Accepted: 11/12/2012] [Indexed: 11/24/2022]
Abstract
Important biological events associated with plasma membranes, such as signal transduction, cell adhesion, and protein trafficking, are mediated through the membrane microdomains. We have developed a novel method termed enzyme-mediated activation of radical sources (EMARS) to identify coclustering molecules on the cell surface under living conditions, which features a radical formation from an aryl azide reagent by horseradish peroxidase (HRP). For identification of molecules labeled by the EMARS reaction, antibody array system and mass spectrometry-based proteomics approaches are available. Spatio- temporally-regulated interaction between β1 integrin and ErbB4 involved in fibronectin-dependent cell migration and therapeutic antibody-stimulated interaction between FGFR3 and CD20 were discovered using the EMARS method.
Collapse
|
30
|
Kolozsvári B, Bakó É, Bécsi B, Kiss A, Czikora Á, Tóth A, Vámosi G, Gergely P, Erdődi F. Calcineurin regulates endothelial barrier function by interaction with and dephosphorylation of myosin phosphatase. Cardiovasc Res 2012; 96:494-503. [PMID: 22869619 DOI: 10.1093/cvr/cvs255] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
AIMS Calcineurin (CN) influences myosin phosphorylation and alters endothelial barrier function; however, the molecular mechanism is still obscure. Here we examine whether CN controls myosin phosphorylation via mediating the phosphorylation state of Thr696 in myosin phosphatase (MP) target subunit 1 (MYPT1), the phosphorylation site inhibitory to the catalytic activity of MP. METHODS AND RESULTS Exposure of bovine or human pulmonary artery endothelial cells (BPAECs or HPAECs) to the CN inhibitor cyclosporin A (CsA) induces a rise in intracellular Ca(2+) and increases the phosphorylation level of cofilin(Ser3) and MYPT1(Thr696) in a Ca(2+)-and Rho-kinase-dependent manner. An active catalytic fragment of CN overexpressed in tsA201 cells decreases endogenous MYPT-phospho-Thr696 (MYPT1(pThr696)) levels. Purified CN dephosphorylates (32)P-labelled MYPT1, suggesting direct action of CN on this substrate. Interaction of MYPT1 with CN is revealed by MYPT1 pull-down experiments and colocalization in both BPAECs and HPAECs as well as by surface plasmon resonance (SPR)-based binding studies. Stabilization of the MYPT1-CN complex occurs via the MYPT1(300PLIEST305) sequence similar to the CN substrate-docking PxIxIT-motif. Thrombin induces a transient increase of MYPT1(pThr696) in BPAECs, whereas its combination with CsA results in maintained phosphorylation levels of both MYPT1(pThr696) and myosin. These phosphorylation events might correlate with changes in endothelial permeability since CsA slows down the recovery from the thrombin-induced decrease of the transendothelial electrical resistance of the BPAEC monolayer. CONCLUSION CN may improve endothelial barrier function via inducing dephosphorylation of cofilin(pSer3) and by interaction with MYPT1 and activating MP through MYPT1(pThr696) dephosphorylation, thereby affecting actin polymerization and decreasing myosin phosphorylation.
Collapse
Affiliation(s)
- Bernadett Kolozsvári
- Department of Medical Chemistry, Medical and Health Science Center, University of Debrecen, Nagyerdei krt 98, Debrecen H-4032, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Boratkó A, Gergely P, Csortos C. Cell cycle dependent association of EBP50 with protein phosphatase 2A in endothelial cells. PLoS One 2012; 7:e35595. [PMID: 22523604 PMCID: PMC3327649 DOI: 10.1371/journal.pone.0035595] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 03/19/2012] [Indexed: 12/19/2022] Open
Abstract
Ezrin-radixin-moesin (ERM)-binding phosphoprotein 50 (EBP50) is a phosphorylatable PDZ domain-containing adaptor protein that is abundantly expressed in epithelium but was not yet studied in the endothelium. We report unusual nuclear localization of EBP50 in bovine pulmonary artery endothelial cells (BPAEC). Immunofluorescent staining and cellular fractionation demonstrated that EBP50 is present in the nuclear and perinuclear region in interphase cells. In the prophase of mitosis EBP50 redistributes to the cytoplasmic region in a phosphorylation dependent manner and during mitosis EBP50 co-localizes with protein phosphatase 2A (PP2A). Furthermore, in vitro wound healing of BPAEC expressing phospho-mimic mutant of EBP50 was accelerated indicating that EBP50 is involved in the regulation of the cell division. Cell cycle dependent specific interactions were detected between EBP50 and the subunits of PP2A (A, C, and Bα) with immunoprecipitation and pull-down experiments. The interaction of EBP50 with the Bα containing form of PP2A suggests that this holoenzyme of PP2A can be responsible for the dephosphorylation of EBP50 in cytokinesis. Moreover, the results underline the significance of EBP50 in cell division via reversible phosphorylation of the protein with cyclin dependent kinase and PP2A in normal cells.
Collapse
Affiliation(s)
- Anita Boratkó
- Department of Medical Chemistry, University of Debrecen Medical and Health Science Center, Debrecen, Hungary
| | - Pál Gergely
- Department of Medical Chemistry, University of Debrecen Medical and Health Science Center, Debrecen, Hungary
- Cell Biology and Signaling Research Group of the Hungarian Academy of Sciences, University of Debrecen Medical and Health Science Center, Debrecen, Hungary
| | - Csilla Csortos
- Department of Medical Chemistry, University of Debrecen Medical and Health Science Center, Debrecen, Hungary
- * E-mail:
| |
Collapse
|
32
|
de Meyer FJM, Rodgers JM, Willems TF, Smit B. Molecular simulation of the effect of cholesterol on lipid-mediated protein-protein interactions. Biophys J 2011; 99:3629-38. [PMID: 21112287 DOI: 10.1016/j.bpj.2010.09.030] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 09/14/2010] [Accepted: 09/16/2010] [Indexed: 12/01/2022] Open
Abstract
Experiments and molecular simulations have shown that the hydrophobic mismatch between proteins and membranes contributes significantly to lipid-mediated protein-protein interactions. In this article, we discuss the effect of cholesterol on lipid-mediated protein-protein interactions as function of hydrophobic mismatch, protein diameter and protein cluster size, lipid tail length, and temperature. To do so, we study a mesoscopic model of a hydrated bilayer containing lipids and cholesterol in which proteins are embedded, with a hybrid dissipative particle dynamics-Monte Carlo method. We propose a mechanism by which cholesterol affects protein interactions: protein-induced, cholesterol-enriched, or cholesterol-depleted lipid shells surrounding the proteins affect the lipid-mediated protein-protein interactions. Our calculations of the potential of mean force between proteins and protein clusters show that the addition of cholesterol dramatically reduces repulsive lipid-mediated interactions between proteins (protein clusters) with positive mismatch, but does not affect attractive interactions between proteins with negative mismatch. Cholesterol has only a modest effect on the repulsive interactions between proteins with different mismatch.
Collapse
Affiliation(s)
- Frédérick J-M de Meyer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California, USA.
| | | | | | | |
Collapse
|
33
|
Duan W, Zhou J, Zhang S, Zhao K, Zhao L, Ogata K, Sakaue T, Mori A, Wei T. ESeroS-GS modulates lipopolysaccharide-induced macrophage activation by impairing the assembly of TLR-4 complexes in lipid rafts. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:772-83. [PMID: 21276822 DOI: 10.1016/j.bbamcr.2011.01.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 01/13/2011] [Accepted: 01/18/2011] [Indexed: 01/29/2023]
Abstract
The binding of lipopolysaccharides (LPS) to macrophages results in inflammatory responses. In extreme cases it can lead to endotoxic shock, often resulting in death. A broad range of antioxidants, including tocopherols, can reduce LPS activity in vitro and in vivo. To elucidate the underlying mechanisms of their action, we investigated the effect of the sodium salt of γ-L-glutamyl-S-[2-[[[3,4-dihydro-2,5,7,8-tetramethyl-2-(4,8,12-trimethyltridecyl)-2H-1-benzopyran-6-yl]oxy]carbonyl]-3-[[2-(1H-indol-3-yl)ethyl]amino]-3-oxopropyl]-L-cysteinylglycine (ESeroS-GS), a novel α-tocopherol derivative, on LPS-induced inflammation in vitro and in vivo. ESeroS-GS reduced the transcription of TNF-α, IL-1β, IL-6 and iNOS genes in a dose-dependent manner in RAW264.7 macrophages, and inhibited the release of these inflammatory factors. In addition, ESeroS-GS inhibited LPS-induced mortality in a mouse sepsis model. Electrophoretic mobility shift assays (EMSA) and reporter gene assays revealed that ESeroS-GS down-regulated the transcriptional activity of NF-κB. By analyzing the partitioning of CD14 and Toll-like receptor 4 (TLR-4) in cell membrane microdomains, we found that ESeroS-GS attenuates the binding of LPS to RAW264.7 cells via interfering with the relocation of CD14 and TLR-4 to lipid rafts, blocking the activation of interleukin-1 receptor-associated kinase 1 (IRAK-1), and inhibiting the consequent phosphorylation of TAK1 and IKKα/β, which together account for the suppression of NF-κB activation. Taken together, our data suggest that ESeroS-GS can modulate LPS signaling in macrophages by impairing TLR-4 complex assembly via a lipid raft dependent mechanism. This article is part of a Special Issue entitled: 11th European Symposium on Calcium.
Collapse
Affiliation(s)
- Wenjuan Duan
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Honke K, Kotani N. The enzyme-mediated activation of radical source reaction: a new approach to identify partners of a given molecule in membrane microdomains. J Neurochem 2011; 116:690-5. [PMID: 21214558 DOI: 10.1111/j.1471-4159.2010.07027.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Important biological events associated with plasma membranes, such as signal transduction, cell adhesion, and protein trafficking, are mediated through the membrane microdomains. However, it is difficult to assess the issue of how they assemble under physiological conditions. We developed a new approach to identify partners of a given molecule on the cell surface in living cells. The important feature of this system, termed as enzyme-mediated activation of radical source, is that activation of cross-linking reagent arylazide-biotin tag can be accomplished not by ultraviolet light, but by an enzyme, horseradish peroxidase. By using this method, we found that many kinds of receptor tyrosine kinases are associated with β1 integrin whereas a few receptor tyrosine kinases are associated with ganglioside GM1 in HeLa S3 cells. This system is a comprehensive approach to identify interactions between cell surface molecules under living conditions. The advantages of this approach are as follows: (i) easy, high throughput, and without the need for special equipment, (ii) applicable to systematic approaches such as proteomic analysis, (iii) applicable to studies on the interactions among not only proteins but also glycans and lipids. The biochemical approach although the enzyme-mediated activation of radical source reaction will provide a new insight into a wide range of research concerning cis-interaction between biomolecules on the cell surface in living cells.
Collapse
Affiliation(s)
- Koichi Honke
- Department of Biochemistry, Kochi University Medical School, Nankoku, Kochi, Japan.
| | | |
Collapse
|
35
|
Vondalova Blanarova O, Jelinkova I, Szoor A, Skender B, Soucek K, Horvath V, Vaculova A, Andera L, Sova P, Szollosi J, Hofmanova J, Vereb G, Kozubik A. Cisplatin and a potent platinum(IV) complex-mediated enhancement of TRAIL-induced cancer cells killing is associated with modulation of upstream events in the extrinsic apoptotic pathway. Carcinogenesis 2010; 32:42-51. [DOI: 10.1093/carcin/bgq220] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
36
|
Núñez E, Alonso-Torres P, Fornés A, Aragón C, López-Corcuera B. The neuronal glycine transporter GLYT2 associates with membrane rafts: functional modulation by lipid environment. J Neurochem 2010; 105:2080-90. [PMID: 18266927 DOI: 10.1111/j.1471-4159.2008.05292.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The neuronal glycine transporter GLYT2 is a plasma membrane protein that removes the neurotransmitter glycine from the synaptic cleft, thereby aiding the pre-synaptic terminal reloading and the termination of the glycinergic signal. Missense mutations in the gene encoding GLYT2 (SLC6A5) cause hyperekplexia in humans. The activity of GLYT2 seems to be highly regulated. In this report, we demonstrate that GLYT2 is associated with membrane rafts in the plasma membrane of brainstem terminals and neurons. The transporter is localized to Triton X-100-insoluble light synaptosomal membranes together with flotillin-1, a marker protein for membrane rafts, in a methyl-beta-cyclodextrin (MbetaCD)-sensitive manner. In brainstem primary neurons, the GLYT2 punctuate pattern visualized by confocal microscopy was modified by cholesterol depletion with MbetaCD, unlike other non-raft neuronal markers. GLYT2-associated gold particles were observed by electron microscopy on purified rafts from brainstem synaptosomes. Furthermore, either in brainstem terminals and cultured neurons, the pharmacological reduction of the levels of raft components, cholesterol and sphingomyelin, impairs both the association of GLYT2 with membrane rafts and its transport activity. Thus, GLYT2 may require membrane raft location for optimal function, and therefore the lipid environment may constitute a new mechanism to modulate GLYT2.
Collapse
Affiliation(s)
- Enrique Núñez
- Departamento de Biología Molecular, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | | | |
Collapse
|
37
|
Rafts and the battleships of defense: The multifaceted microdomains for positive and negative signals in immune cells. Immunol Lett 2010; 130:2-12. [DOI: 10.1016/j.imlet.2009.12.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Revised: 12/13/2009] [Accepted: 12/13/2009] [Indexed: 11/20/2022]
|
38
|
Orsini F, Santacroce M, Arosio P, Sacchi VF. Observing Xenopus laevis oocyte plasma membrane by atomic force microscopy. Methods 2010; 51:106-13. [DOI: 10.1016/j.ymeth.2009.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Revised: 12/02/2009] [Accepted: 12/02/2009] [Indexed: 10/20/2022] Open
|
39
|
Jenei A, Kormos J, Szentesi G, Veres AJ, Varga S, Bodnár A, Damjanovich S, Mátyus L. Non-random distribution of interleukin receptors on the cell surface. Chemphyschem 2009; 10:1577-85. [PMID: 19514033 DOI: 10.1002/cphc.200900242] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Spatial organization of cell surface proteins plays a key role in the process of transmembrane signalling. Receptor clustering and changes in their cell surface distribution are often determining factors in the final outcome of ligand-receptor interactions. There are several techniques for assessing the distribution of protein molecules. Fluorescence resonance energy transfer (FRET) is an excellent tool for determining distance relationships of cell surface molecules. However, it does not provide information on the distribution of molecular clusters. Different kinds of microscopies fill this gap. The evaluation of the images provided by the listed techniques is often questionable. Herein we show the applicability of Ripley's K(t) function as a tool for analyzing the cell surface receptor patterns (Y. Nakamura, et al., Nature 1994, 369, 330-333). We have implemented an effective image processing algorithm for fast localization of gold labels on biological samples. We investigated spatial organization of Interleukin-2R alpha and -15R alpha (IL-2R alpha and IL-15R alpha) on a human CD4+leukaemia T-cell line, Kit225 FT7.10 by using transmission electron microscopy (TEM). TEM analysis showed co-clustering of the two types of alpha-chains even on the few-hundred-nanometer scale. The analysis of our data may contribute to our understanding the action of the IL-2/IL-15 receptor system in T-cell function.
Collapse
Affiliation(s)
- Attila Jenei
- Department of Biophysics and Cell Biology, University of Debrecen, 98 Nagyerdei krt. H-4032 Debrecen, Hungary.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Xu S, Huo J, Gunawan M, Su IH, Lam KP. Activated dectin-1 localizes to lipid raft microdomains for signaling and activation of phagocytosis and cytokine production in dendritic cells. J Biol Chem 2009; 284:22005-22011. [PMID: 19525229 DOI: 10.1074/jbc.m109.009076] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Lipid rafts are plasma membrane microdomains that are enriched in cholesterol, glycosphingolipids, and glycosylphosphatidylinositol-anchored proteins and play an important role in the signaling of ITAM-bearing lymphocyte antigen receptors. Dectin-1 is a C-type lectin receptor (CLR) that recognizes beta-glucan in the cell walls of fungi and triggers signal transduction via its cytoplasmic hemi-ITAM. However, it is not known if similar to antigen receptors, Dectin-1 would also signal via lipid rafts and if the integrity of lipid raft microdomains is important for the physiological functions mediated by Dectin-1. We demonstrate here using sucrose gradient ultracentrifugation and confocal microscopy that Dectin-1 translocates to lipid rafts upon stimulation of dendritic cells (DCs) with the yeast derivative zymosan or beta-glucan. In addition, two key signaling molecules, Syk and PLCgamma2 are also recruited to lipid rafts upon the activation of Dectin-1, suggesting that lipid raft microdomains facilitate Dectin-1 signaling. Disruption of lipid raft integrity with the synthetic drug, methyl-beta-cyclodextrin (betamD) leads to reduced intracellular Ca2+ flux and defective Syk and ERK phosphorylation in Dectin-1-activated DCs. Furthermore, betamD-treated DCs have significantly attenuated production of IL-2, IL-10, and TNFalpha upon Dectin-1 engagement, and they also exhibit impaired phagocytosis of zymosan particles. Taken together, the data indicate that Dectin-1 and perhaps also other CLRs are recruited to lipid rafts upon activation and that the integrity of lipid rafts is important for the signaling and cellular functions initiated by this class of innate receptors.
Collapse
Affiliation(s)
- Shengli Xu
- Laboratory of Immunology, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore 138668
| | - Jianxin Huo
- Laboratory of Immunology, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore 138668
| | - Merry Gunawan
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - I-Hsin Su
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Kong-Peng Lam
- Laboratory of Immunology, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore 138668; Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074
| |
Collapse
|
41
|
Kiss E, Nagy P, Balogh A, Szöllosi J, Matkó J. Cytometry of raft and caveola membrane microdomains: from flow and imaging techniques to high throughput screening assays. Cytometry A 2008; 73:599-614. [PMID: 18473380 DOI: 10.1002/cyto.a.20572] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The evolutionarily developed microdomain structure of biological membranes has gained more and more attention in the past decade. The caveolin-free "membrane rafts," the caveolin-expressing rafts (caveolae), as well as other membrane microdomains seem to play an essential role in controlling and coordinating cell-surface molecular recognition, internalization/endocytosis of the bound molecules or pathogenic organisms and in regulation of transmembrane signal transduction processes. Therefore, in many research fields (e.g. neurobiology and immunology), there is an ongoing need to understand the nature of these microdomains and to quantitatively characterize their lipid and protein composition under various physiological and pathological conditions. Flow and image cytometry offer many sophisticated and routine tools to study these questions. In this review, we give an overview of the past efforts to detect and characterize these membrane microdomains by the use of classical cytometric technologies, and finally we will discuss the results and perspectives of a new line of raft cytometry, the "high throughput screening assays of membrane microdomains," based on "lipidomic" and "proteomic" approaches.
Collapse
Affiliation(s)
- Endre Kiss
- Immunology Research Group of the Hungarian Academy of Sciences at Eötvös Loránd University, Budapest, Hungary
| | | | | | | | | |
Collapse
|
42
|
de Bakker BI, Bodnár A, van Dijk EMHP, Vámosi G, Damjanovich S, Waldmann TA, van Hulst NF, Jenei A, Garcia-Parajo MF. Nanometer-scale organization of the alpha subunits of the receptors for IL2 and IL15 in human T lymphoma cells. J Cell Sci 2008; 121:627-33. [PMID: 18287585 DOI: 10.1242/jcs.019513] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Interleukin 2 and interleukin 15 (IL2 and IL15, respectively) provide quite distinct contributions to T-cell-mediated immunity, despite having similar receptor composition and signaling machinery. As most of the proposed mechanisms underlying this apparent paradox attribute key significance to the individual alpha-chains of IL2 and IL15 receptors, we investigated the spatial organization of the receptors IL2Ralpha and IL15Ralpha at the nanometer scale expressed on a human CD4+ leukemia T cell line using single-molecule-sensitive near-field scanning optical microscopy (NSOM). In agreement with previous findings, we here confirm clustering of IL2Ralpha and IL15Ralpha at the submicron scale. In addition to clustering, our single-molecule data reveal that a non-negligible percentage of the receptors are organized as monomers. Only a minor fraction of IL2Ralpha molecules reside outside the clustered domains, whereas approximately 30% of IL15Ralpha molecules organize as monomers or small clusters, excluded from the main domain regions. Interestingly, we also found that the packing densities per unit area of both IL2Ralpha and IL15Ralpha domains remained constant, suggesting a 'building block' type of assembly involving repeated structures and composition. Finally, dual-color NSOM demonstrated co-clustering of the two alpha-chains. Our results should aid understanding the action of the IL2R-IL15R system in T cell function and also might contribute to the more rationale design of IL2R- or IL15R-targeted immunotherapy agents for treating human leukemia.
Collapse
Affiliation(s)
- Bärbel I de Bakker
- Applied Optics group, Faculty of Science and Technology, MESA+ Research Institute for Nanotechnology, University of Twente, PO Box 217, 7500 AE Enschede, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Biochemical visualization of cell surface molecular clustering in living cells. Proc Natl Acad Sci U S A 2008; 105:7405-9. [PMID: 18495923 DOI: 10.1073/pnas.0710346105] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Many plasma membrane-resident molecules cluster with other molecules to collaborate in a variety of biological events. We herein report a sensitive and simple method to identify components of cell surface molecular clusters in living cells. This method includes a recently established reaction, called the enzyme-mediated activation of radical source (EMARS), to label molecules within a limited distance ( approximately 200-300 nm) from the probed molecule on which HRP is set. Because the size of this active area is close to that of the reported membrane clusters, it is suggested that the labeled molecules cluster with the probed molecule in the same membrane domain. A combination of the EMARS reaction and antibody array analysis demonstrated that many kinds of receptor tyrosine kinases (RTKs) formed clusters with beta1 integrin in HeLa S3 cells. A similar antibody array analysis after the EMARS reaction with three HRP-labeled antibodies against growth factor receptors showed the patterns of biotinylated RTKs to be substantially different from each other. These results suggest that different types of cell surface molecular clusters can thus be distinguished using the EMARS reaction. Therefore, the present "biochemical visualization" method is expected to be a powerful tool to elucidate molecular clustering on the cell surface of living cells in various contexts.
Collapse
|
44
|
Jahn KA, Braet F. Monitoring membrane rafts in colorectal cancer cells by means of correlative fluorescence electron microscopy (CFEM). Micron 2008; 39:1393-7. [PMID: 18495485 DOI: 10.1016/j.micron.2008.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2008] [Revised: 04/10/2008] [Accepted: 04/10/2008] [Indexed: 12/20/2022]
Abstract
Detergent-resistant membrane (DRM) rafts have been shown to play a pivotal role in regulating key cell biological processes, such as signal transduction, cellular transport and cell survival. The fine structure of membrane rafts are studied using various different imaging approaches and the outcomes are largely dependent on the detection methodology applied. All these microscopy techniques which employ light-, laser- and photon-optics, electrons as well as atomic force probing are characterized on their turn by their strengths and limitations for membrane raft identification. This explains in part the diversity of definitions available to describe these peculiar membrane structures. We present herewith an alternative and uncomplicated microscopy tool to study fluorescently labelled DRMs with information at the transmission electron microscopical level of the same cell, enabling us to obtain a snapshot of the morpho-functional relationships between the cell's interior and DRMs. The proposed approach of correlative fluorescence electron microscopy (CFEM) can therefore be considered as an additional alternative imaging approach to unravel DRM structure-function relationships from micro- to nanometre length scales, from the cell to the molecule.
Collapse
Affiliation(s)
- Kristina A Jahn
- Australian Key Centre for Microscopy and Microanalysis, Madsen Building F09, The University of Sydney, NSW 2006, Australia
| | | |
Collapse
|
45
|
Tanikawa R, Tanikawa T, Okada Y, Nakano K, Hirashima M, Yamauchi A, Hosokawa R, Tanaka Y. Interaction of galectin-9 with lipid rafts induces osteoblast proliferation through the c-Src/ERK signaling pathway. J Bone Miner Res 2008; 23:278-86. [PMID: 17907924 DOI: 10.1359/jbmr.071008] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
UNLABELLED Galectin-9 is a beta-galactoside-binding lectin expressed in various tissues, including bone. The role of galectin-9 in human osteoblasts, however, remains unclear. This study showed that galectin-9 interacts with lipid rafts and induces osteoblast proliferation through the c-Src/ERK signaling pathway. INTRODUCTION Galectin-9 is a beta-galactoside-binding lectin that modulates many biological functions by interacting with particular carbohydrates attached to proteins and lipids. However, the role of galectin-9 in bone metabolism and osteoblast proliferation remains unclear. This study investigated the effects of galectin-9 on osteoblast proliferation and its signaling mechanisms. MATERIALS AND METHODS The effect of galectin-9 on osteoblast proliferation was tested by measuring the conversion of tetrazolium salt WST-8 to formazan. Protein phosphorylation was assayed by western blotting and confocal microscopy was used to localize lipid rafts. RESULTS Galectin-9-induced proliferation of the obtained osteoblasts in a dose-dependent manner, whereas galectin-1, -3, and -4 did not. Galectin-9-induced phosphorylation of c-Src and subsequent ERK1/ERK2 in the osteoblasts. The galectin-9-induced phosphorylation and proliferation were inhibited by PP2, a selective inhibitor of c-Src. Galectin-9-induced clustering of lipid rafts detected by cholera toxin B (CTB; binding the raft-resident ganglioside GM1) using confocal microscopy. Cross-linking of the GM1 ganglioside with CTB by anti-CTB antibody-induced phosphorylation of c-Src, whereas disruption of galectin-9-induced lipid rafts by beta-methylcyclodextrin reduced c-Src phosphorylation and proliferation of the cells. CONCLUSIONS These results suggest that galectin-9, but not other galectins, induced proliferation of human osteoblasts through clustering lipid rafts on membrane and subsequent phosphorylation of the c-Src/ERK signaling pathway.
Collapse
Affiliation(s)
- Rena Tanikawa
- First Department of Internal Medicine, University of Occupational and Environmental Health, School of Medicine, Kitakyushu, Japan
| | | | | | | | | | | | | | | |
Collapse
|
46
|
A biophysical approach to IL-2 and IL-15 receptor function: localization, conformation and interactions. Immunol Lett 2008; 116:117-25. [PMID: 18280585 DOI: 10.1016/j.imlet.2007.12.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2007] [Accepted: 12/30/2007] [Indexed: 12/25/2022]
Abstract
Interleukin-2 and interleukin-15 (IL-2, IL-15) are key participants in T and NK cell activation and function. Sharing the beta and gamma receptor subunits results in several common functions: e.g. the promotion of T cell proliferation. On the other hand, due to their distinct alpha receptor subunits, they also play opposing roles in immune processes such as activation induced cell death and immunological memory. Divergence of signaling pathways must ensue already at the plasma membrane where the cytokines interact with their receptors. Therefore understanding molecular details of receptor organization and mapping interactions with other membrane proteins that might influence receptor conformation and function, are of key importance. Biophysical/advanced microscopic methods (fluorescence resonance energy transfer (FRET), fluorescence crosscorrelation spectroscopy (FCCS), near-field scanning optical microscopy (NSOM), X-ray crystallography, surface plasmon resonance, NMR spectroscopy) have been instrumental in clarifying the details of receptor structure and organization from the atomic level to the assembly and dynamics of supramolecular clusters. In this short review some important contributions shaping our current view of IL-2 and IL-15 receptors are presented.
Collapse
|
47
|
Bene L, Kanyári Z, Bodnár A, Kappelmayer J, Waldmann TA, Vámosi G, Damjanovich L. Colorectal carcinoma rearranges cell surface protein topology and density in CD4+ T cells. Biochem Biophys Res Commun 2007; 361:202-7. [PMID: 17658476 DOI: 10.1016/j.bbrc.2007.07.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2007] [Accepted: 07/05/2007] [Indexed: 11/24/2022]
Abstract
Previously, we described conserved protein clusters including MHC I and II glycoproteins, ICAM-1 adhesion molecules, and interleukin-2 and -15 receptors in lipid rafts of several human cell types. Differential protein-protein interactions can modulate function, thus influence cell fate. Therefore, we analyzed supramolecular clusters of CD4(+) T cells from draining lymph nodes and peripheral blood of colorectal carcinoma patients, and compared these to healthy controls. Superclusters of MHC I and II with IL-2/15 receptors were identified by confocal microscopy on all cell types. Flow-cytometric FRET revealed molecular associations of these proteins with each other and with ICAM-1 as well. In draining lymph nodes expression levels of all these proteins were lower, and interactions, particularly between IL-2/15 receptors and MHC molecules weakened or disappeared as compared to the control. Stimuli/local conditions can rearrange cell surface protein patterns on the same cell type in the same patient, having important implications on further function and cell fate.
Collapse
Affiliation(s)
- László Bene
- Department of Biophysics and Cell Biology, Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | | | | | | | | | | | | |
Collapse
|
48
|
Field EH, Kulhankova K, Nasr ME. Natural Tregs, CD4+CD25+ inhibitory hybridomas, and their cell contact dependent suppression. Immunol Res 2007; 39:62-78. [DOI: 10.1007/s12026-007-0064-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 11/30/2022]
|
49
|
Drbal K, Moertelmaier M, Holzhauser C, Muhammad A, Fuertbauer E, Howorka S, Hinterberger M, Stockinger H, Schütz GJ. Single-molecule microscopy reveals heterogeneous dynamics of lipid raft components upon TCR engagement. Int Immunol 2007; 19:675-84. [PMID: 17446208 DOI: 10.1093/intimm/dxm032] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The existence of lipid rafts and their importance for immunoreceptor signaling is highly debated. By non-invasive single molecule imaging, we analyzed the dynamics of the T-cell antigen receptor (TCR), the lipid raft-associated glycosylphosphatidylinositol (GPI) proteins CD48 and CD59 and the major leukocyte phosphatase CD45 in living naive T lymphocytes. TCR triggering induced the immobilization of CD45 and CD48 at different positions within the T-cell interface. The second GPI protein, CD59, did not co-immobilize indicating lipid raft heterogeneity in living T lymphocytes. A novel biochemical approach confirmed that lipid raft components are not associated in the plasma membrane of resting cells, and variably associate with specific receptors to distinct lipid rafts upon activation.
Collapse
Affiliation(s)
- Karel Drbal
- Competence Centre for Biomolecular Therapeutics Research Vienna, A-1090, Vienna
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Takemura Y, Ouchi N, Shibata R, Aprahamian T, Kirber MT, Summer RS, Kihara S, Walsh K. Adiponectin modulates inflammatory reactions via calreticulin receptor-dependent clearance of early apoptotic bodies. J Clin Invest 2007; 117:375-86. [PMID: 17256056 PMCID: PMC1770947 DOI: 10.1172/jci29709] [Citation(s) in RCA: 273] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Accepted: 12/05/2006] [Indexed: 02/06/2023] Open
Abstract
Obesity and type 2 diabetes are associated with chronic inflammation. Adiponectin is an adipocyte-derived hormone with antidiabetic and antiinflammatory actions. Here, we demonstrate what we believe to be a previously undocumented activity of adiponectin, facilitating the uptake of early apoptotic cells by macrophages, an essential feature of immune system function. Adiponectin-deficient (APN-KO) mice were impaired in their ability to clear apoptotic thymocytes in response to dexamethasone treatment, and these animals displayed a reduced ability to clear early apoptotic cells that were injected into their intraperitoneal cavities. Conversely, adiponectin administration promoted the clearance of apoptotic cells by macrophages in both APN-KO and wild-type mice. Adiponectin overexpression also promoted apoptotic cell clearance and reduced features of autoimmunity in lpr mice whereas adiponectin deficiency in lpr mice led to a further reduction in apoptotic cell clearance, which was accompanied by exacerbated systemic inflammation. Adiponectin was capable of opsonizing apoptotic cells, and phagocytosis of cell corpses was mediated by the binding of adiponectin to calreticulin on the macrophage cell surface. We propose that adiponectin protects the organism from systemic inflammation by promoting the clearance of early apoptotic cells by macrophages through a receptor-dependent pathway involving calreticulin.
Collapse
Affiliation(s)
- Yukihiro Takemura
- Molecular Cardiology Unit, Whitaker Cardiovascular Institute,
Section of Molecular Medicine, and
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA.
Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Noriyuki Ouchi
- Molecular Cardiology Unit, Whitaker Cardiovascular Institute,
Section of Molecular Medicine, and
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA.
Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Rei Shibata
- Molecular Cardiology Unit, Whitaker Cardiovascular Institute,
Section of Molecular Medicine, and
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA.
Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tamar Aprahamian
- Molecular Cardiology Unit, Whitaker Cardiovascular Institute,
Section of Molecular Medicine, and
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA.
Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Michael T. Kirber
- Molecular Cardiology Unit, Whitaker Cardiovascular Institute,
Section of Molecular Medicine, and
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA.
Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ross S. Summer
- Molecular Cardiology Unit, Whitaker Cardiovascular Institute,
Section of Molecular Medicine, and
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA.
Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shinji Kihara
- Molecular Cardiology Unit, Whitaker Cardiovascular Institute,
Section of Molecular Medicine, and
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA.
Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kenneth Walsh
- Molecular Cardiology Unit, Whitaker Cardiovascular Institute,
Section of Molecular Medicine, and
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA.
Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|