1
|
Redpath G, Deo N. Serotonin: an overlooked regulator of endocytosis and endosomal sorting? Biol Open 2022; 11:bio059057. [PMID: 35076063 PMCID: PMC8801889 DOI: 10.1242/bio.059057] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/09/2021] [Indexed: 12/23/2022] Open
Abstract
Serotonin is a neurotransmitter and a hormone that is typically associated with regulating our mood. However, the serotonin transporter and receptors are expressed throughout the body, highlighting the much broader, systemic role of serotonin in regulating human physiology. A substantial body of data strongly implicates serotonin as a fundamental regulator of endocytosis and endocytic sorting. Serotonin has the potential to enhance endocytosis through three distinct mechanisms - serotonin signalling, serotonylation and insertion into the plasma membrane - although the interplay and relationship between these mechanisms has not yet been explored. Endocytosis is central to the cellular response to the extracellular environment, controlling receptor distribution on the plasma membrane to modulate signalling, neurotransmitter release and uptake, circulating protein and lipid cargo uptake, and amino acid internalisation for cell proliferation. Uncovering the range of cellular and physiological circumstances in which serotonin regulates endocytosis is of great interest for our understanding of how serotonin regulates mood, and also the fundamental understanding of endocytosis and its regulation throughout the body. This article has an associated Future Leader to Watch interview with the first author of the paper.
Collapse
Affiliation(s)
- Gregory Redpath
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences and the ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney 2052, Australia
| | - Nikita Deo
- Department of Biochemistry, University of Otago, Dunedin 9016, New Zealand
| |
Collapse
|
2
|
Macias A, de la Cruz A, Peraza DA, de Benito-Bueno A, Gonzalez T, Valenzuela C. K V1.5-K Vβ1.3 Recycling Is PKC-Dependent. Int J Mol Sci 2021; 22:ijms22031336. [PMID: 33572906 PMCID: PMC7866247 DOI: 10.3390/ijms22031336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/12/2021] [Accepted: 01/26/2021] [Indexed: 11/26/2022] Open
Abstract
KV1.5 channel function is modified by different regulatory subunits. KVβ1.3 subunits assemble with KV1.5 channels and induce a fast and incomplete inactivation. Inhibition of PKC abolishes the KVβ1.3-induced fast inactivation, decreases the amplitude of the current KV1.5–KVβ1.3 and modifies their pharmacology likely due to changes in the traffic of KV1.5–KVβ1.3 channels in a PKC-dependent manner. In order to analyze this hypothesis, HEK293 cells were transfected with KV1.5–KVβ1.3 channels, and currents were recorded by whole-cell configuration of the patch-clamp technique. The presence of KV1.5 in the membrane was analyzed by biotinylation techniques, live cell imaging and confocal microscopy approaches. PKC inhibition resulted in a decrease of 33 ± 7% of channels in the cell surface due to reduced recycling to the plasma membrane, as was confirmed by confocal microscopy. Live cell imaging indicated that PKC inhibition almost abolished the recycling of the KV1.5–KVβ1.3 channels, generating an accumulation of channels into the cytoplasm. All these results suggest that the trafficking regulation of KV1.5–KVβ1.3 channels is dependent on phosphorylation by PKC and, therefore, they could represent a clinically relevant issue, mainly in those diseases that exhibit modifications in PKC activity.
Collapse
Affiliation(s)
- Alvaro Macias
- Instituto de Investigaciones Biomédicas Madrid CSIC-UAM. C/Arturo Duperier 4, 28029 Madrid, Spain; (A.d.l.C.); (D.A.P.); (A.d.B.-B.); (T.G.)
- CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Correspondence: (A.M.); (C.V.); Tel.: +34-91-453-1200 (A.M.); +34-91-585-4493 (C.V.)
| | - Alicia de la Cruz
- Instituto de Investigaciones Biomédicas Madrid CSIC-UAM. C/Arturo Duperier 4, 28029 Madrid, Spain; (A.d.l.C.); (D.A.P.); (A.d.B.-B.); (T.G.)
| | - Diego A. Peraza
- Instituto de Investigaciones Biomédicas Madrid CSIC-UAM. C/Arturo Duperier 4, 28029 Madrid, Spain; (A.d.l.C.); (D.A.P.); (A.d.B.-B.); (T.G.)
- CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Angela de Benito-Bueno
- Instituto de Investigaciones Biomédicas Madrid CSIC-UAM. C/Arturo Duperier 4, 28029 Madrid, Spain; (A.d.l.C.); (D.A.P.); (A.d.B.-B.); (T.G.)
- CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Teresa Gonzalez
- Instituto de Investigaciones Biomédicas Madrid CSIC-UAM. C/Arturo Duperier 4, 28029 Madrid, Spain; (A.d.l.C.); (D.A.P.); (A.d.B.-B.); (T.G.)
- CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), 28029 Madrid, Spain
| | - Carmen Valenzuela
- Instituto de Investigaciones Biomédicas Madrid CSIC-UAM. C/Arturo Duperier 4, 28029 Madrid, Spain; (A.d.l.C.); (D.A.P.); (A.d.B.-B.); (T.G.)
- CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Correspondence: (A.M.); (C.V.); Tel.: +34-91-453-1200 (A.M.); +34-91-585-4493 (C.V.)
| |
Collapse
|
3
|
Salama MF, Liu M, Clarke CJ, Espaillat MP, Haley JD, Jin T, Wang D, Obeid LM, Hannun YA. PKCα is required for Akt-mTORC1 activation in non-small cell lung carcinoma (NSCLC) with EGFR mutation. Oncogene 2019; 38:7311-7328. [PMID: 31420605 PMCID: PMC6883150 DOI: 10.1038/s41388-019-0950-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 12/29/2022]
Abstract
Mutational activation of the epidermal growth factor receptor (EGFR) is a major player in the pathogenesis of non-small cell lung cancer (NSCLC). NSCLC patients with constitutively active EGFR mutations eventually develop drug resistance against EGFR tyrosine-kinase inhibitors; therefore, better understandings of key components of mutant EGFR (mtEGFR) signaling are required. Here, we initially observed aberrantly high expression of protein kinase Cα (PKCα) in lung adenocarcinomas, especially those with EGFR mutations, and proceeded to examine the role of PKCα in the regulation of the signaling pathways downstream of mtEGFR. The results showed that NSCLC cell lines with constitutively active EGFR mutations tend to have very or moderately high PKCα levels. Furthermore, PKCα was constitutively activated in HCC827 and H4006 cells which have an EGFR deletion mutation in exon 19. Interestingly, mtEGFR was not required for the induction of PKCα at protein and message levels suggesting that the increased levels of PKCα are due to independent selection. On the other hand, mtEGFR activity was required for robust activation of PKCα. Loss of functions studies revealed that the NSCLC cells rely heavily on PKCα for the activation of the mTORC1 signaling pathway. Unexpectedly, the results demonstrated that PKCα was required for activation of Akt upstream of mTOR but only in cells with the mtEGFR and with the increased expression of PKCα. Functionally, inhibition of PKCα in HCC827 led to caspase-3-dependent apoptosis and a significant decrease in cell survival in response to cellular stress induced by serum starvation. In summary, the results identified important roles of PKCα in regulating mTORC1 activity in lung cancer cells, whereby a primary switching occurs from PKCα-independent to PKCα-dependent signaling in the presence of EGFR mutations. The results present PKCα as a potential synergistic target of personalized treatment for NSCLC with constitutively active mutant forms of EGFR and constitutively active PKCα.
Collapse
Affiliation(s)
- Mohamed F Salama
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
- Stony Brook Cancer Center, Stony Brook University Hospital, Stony Brook, NY, 11794, USA
- Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Mengling Liu
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
- Stony Brook Cancer Center, Stony Brook University Hospital, Stony Brook, NY, 11794, USA
- Department of Immuno-Oncology, HD Biosciences Inc, San Diego, CA, 92121, USA
| | - Christopher J Clarke
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
- Stony Brook Cancer Center, Stony Brook University Hospital, Stony Brook, NY, 11794, USA
| | - Mel Pilar Espaillat
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
- Stony Brook Cancer Center, Stony Brook University Hospital, Stony Brook, NY, 11794, USA
| | - John D Haley
- Departments of Biochemistry and Pathology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Ting Jin
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Daifeng Wang
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY, 11794, USA
- Department of Biostatistics and Medical Informatics and Waisman Center, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Lina M Obeid
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
- Stony Brook Cancer Center, Stony Brook University Hospital, Stony Brook, NY, 11794, USA
| | - Yusuf A Hannun
- Department of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA.
- Stony Brook Cancer Center, Stony Brook University Hospital, Stony Brook, NY, 11794, USA.
- Departments of Biochemistry and Pathology, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
4
|
Dietrich M, Malik MS, Skeie M, Bertelsen V, Stang E. Protein kinase C regulates ErbB3 turnover. Exp Cell Res 2019; 382:111473. [PMID: 31233741 DOI: 10.1016/j.yexcr.2019.06.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 01/07/2023]
Abstract
ErbB3, which belongs to the epidermal growth factor receptor (EGFR) or ErbB family of receptor tyrosine kinases, is involved in progression of several human cancers and a tight regulation of its expression is crucial. An important mechanism for regulation of ErbB proteins is endocytosis and we recently showed that ErbB3, contrary to other ErbB proteins, like EGFR and ErbB2, is constitutively internalized and degraded. Several studies show that protein kinase C (PKC) can regulate the activation, localization and stability of EGFR and ErbB2. Activation of PKC causes their down-regulation from the plasma membrane, but instead of being degraded the receptors accumulate in an endosomal recycling compartment. Since little is known about possible connections between ErbB3 and PKC, we have in the present study investigated effects PKC activity has on ErbB3 stability and intracellular trafficking. While PKC inhibition tends to increase ErbB3 degradation, activation of PKC causes ErbB3 stabilization. The stabilization was not due to inhibited internalization, on the contrary we find that expression of ErbB3 at the plasma membrane is reduced upon PMA-induced PKC activation. However, while endocytosed ErbB3 under normal conditions and upon PKC inhibition is found in early endosomal antigen 1 (EEA1) positive early endosomes and lysosomal-associated membrane protein 1 (LAMP1) positive late endosomes/lysosomes, indicating that it follows the classic degradative pathway, ErbB3 localizes to EEA1 and LAMP1 negative compartments upon PMA-induced activation of PKC. Altogether this shows that PKC regulates the stability of ErbB3, and knockdown experiments show that PKCδ is essential in this process. A likely explanation is that PKC regulates endosomal sorting of ErbB3 and that activated PKC sorts ErbB3 away from the degradative pathway.
Collapse
Affiliation(s)
- Markus Dietrich
- Department of Pathology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Marianne Skeie
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Espen Stang
- Department of Pathology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
5
|
Bumpus T, Baskin JM. Clickable Substrate Mimics Enable Imaging of Phospholipase D Activity. ACS CENTRAL SCIENCE 2017; 3:1070-1077. [PMID: 29104923 PMCID: PMC5658752 DOI: 10.1021/acscentsci.7b00222] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Indexed: 05/15/2023]
Abstract
Chemical imaging techniques have played instrumental roles in dissecting the spatiotemporal regulation of signal transduction pathways. Phospholipase D (PLD) enzymes affect cell signaling by producing the pleiotropic lipid second messenger phosphatidic acid via hydrolysis of phosphatidylcholine. It remains a mystery how this one lipid signal can cause such diverse physiological and pathological signaling outcomes, due in large part to a lack of suitable tools for visualizing the spatial and temporal dynamics of its production within cells. Here, we report a chemical method for imaging phosphatidic acid synthesis by PLD enzymes in live cells. Our approach capitalizes upon the enzymatic promiscuity of PLDs, which we show can accept azidoalcohols as reporters in a transphosphatidylation reaction. The resultant azidolipids are then fluorescently tagged using the strain-promoted azide-alkyne cycloaddition, enabling visualization of cellular membranes bearing active PLD enzymes. Our method, termed IMPACT (Imaging Phospholipase D Activity with Clickable Alcohols via Transphosphatidylation), reveals pools of basal and stimulated PLD activities in expected and unexpected locations. As well, we reveal a striking heterogeneity in PLD activities at both the cellular and subcellular levels. Collectively, our studies highlight the importance of using chemical tools to directly visualize, with high spatial and temporal resolution, the subset of signaling enzymes that are active.
Collapse
Affiliation(s)
- Timothy
W. Bumpus
- Department of Chemistry and
Chemical Biology and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, United States
| | - Jeremy M. Baskin
- Department of Chemistry and
Chemical Biology and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
6
|
Liu M, Clarke CJ, Salama MF, Choi YJ, Obeid LM, Hannun YA. Co-ordinated activation of classical and novel PKC isoforms is required for PMA-induced mTORC1 activation. PLoS One 2017; 12:e0184818. [PMID: 28926616 PMCID: PMC5604983 DOI: 10.1371/journal.pone.0184818] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 08/31/2017] [Indexed: 01/19/2023] Open
Abstract
Protein kinase C (PKC) has been shown to activate the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway, a central hub in the regulation of cell metabolism, growth and proliferation. However, the mechanisms by which PKCs activate mTORC1 are still ambiguous. Our previous study revealed that activation of classical PKCs (cPKC) results in the perinuclear accumulation of cPKC and phospholipase D2 (PLD2) in recycling endosomes in a PLD2-dependent manner. Here, we report that mTORC1 activation by phorbol 12,13-myristate acetate (PMA) requires both classic, cPKC, and novel PKC (nPKC) isoforms, specifically PKCη, acting through distinct pathways. The translocation of mTOR to perinuclear lysosomes was detected after treatment of PKC activators, which was not colocalized with PKCα- or RAB11-positive endosomes and was not inhibited by PLD inhibitors. We found that PKCη inhibition by siRNA or bisindolylmaleimide I effectively decreased mTOR accumulation in lysosomes and its activity. Also, we identified that PKCη plays a role upstream of the v-ATPase/Ragulator/Rag pathway in response to PMA. These data provides a spatial aspect to the regulation of mTORC1 by sustained activation of PKC, requiring co-ordinated activation of two distinct elements, the perinuclear accumulation of cPKC- and PLD-containing endosomes and the nPKC-dependent translation of of mTOR in the perinuclear lysosomes. The close proximity of these two distinct compartments shown in this study suggests the possibility that transcompartment signaling may be a factor in the regulation of mTORC1 activity and also underscores the importance of PKCη as a potential therapeutic target of mTORC-related disorders.
Collapse
Affiliation(s)
- Mengling Liu
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States of America
- Stony Brook Cancer Center, Stony Brook University Hospital, Stony Brook, NY, United States of America
| | - Christopher J. Clarke
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States of America
- Stony Brook Cancer Center, Stony Brook University Hospital, Stony Brook, NY, United States of America
| | - Mohamed F. Salama
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States of America
- Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Yeon Ja Choi
- Stony Brook Cancer Center, Stony Brook University Hospital, Stony Brook, NY, United States of America
- Department of Pathology, Stony Brook University, Stony Brook, NY, United States of America
| | - Lina M. Obeid
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States of America
- Stony Brook Cancer Center, Stony Brook University Hospital, Stony Brook, NY, United States of America
| | - Yusuf A. Hannun
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States of America
- Stony Brook Cancer Center, Stony Brook University Hospital, Stony Brook, NY, United States of America
- Department of Pathology, Stony Brook University, Stony Brook, NY, United States of America
| |
Collapse
|
7
|
Zhai X, Leo MD, Jaggar JH. Endothelin-1 Stimulates Vasoconstriction Through Rab11A Serine 177 Phosphorylation. Circ Res 2017; 121:650-661. [PMID: 28696251 DOI: 10.1161/circresaha.117.311102] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/06/2017] [Accepted: 07/07/2017] [Indexed: 01/18/2023]
Abstract
RATIONALE Large-conductance calcium-activated potassium channels (BK) are composed of pore-forming BKα and auxiliary β1 subunits in arterial smooth muscle cells (myocytes). Vasoconstrictors, including endothelin-1 (ET-1), inhibit myocyte BK channels, leading to contraction, but mechanisms involved are unclear. Recent evidence indicates that BKα is primarily plasma membrane localized, whereas the cellular location of β1 can be rapidly altered by Rab11A-positive recycling endosomes. Whether vasoconstrictors regulate the multisubunit composition of surface BK channels to stimulate contraction is unclear. OBJECTIVE Test the hypothesis that ET-1 inhibits BK channels by altering BKα and β1 surface trafficking in myocytes, identify mechanisms involved, and determine functional significance in myocytes of small cerebral arteries. METHODS AND RESULTS ET-1, through activation of PKC (protein kinase C), reduced surface β1 abundance and the proximity of β1 to surface BKα in myocytes. In contrast, ET-1 did not alter surface BKα, total β1, or total BKα proteins. ET-1 stimulated Rab11A phosphorylation, which reduced Rab11A activity. Rab11A serine 177 was identified as a high-probability PKC phosphorylation site. Expression of a phosphorylation-incapable Rab11A construct (Rab11A S177A) blocked the ET-1-induced Rab11A phosphorylation, reduction in Rab11A activity, and decrease in surface β1 protein. ET-1 inhibited single BK channels and transient BK currents in myocytes and stimulated vasoconstriction via a PKC-dependent mechanism that required Rab11A S177. In contrast, NO-induced Rab11A activation, surface trafficking of β1 subunits, BK channel and transient BK current activation, and vasodilation did not involve Rab11A S177. CONCLUSIONS ET-1 stimulates PKC-mediated phosphorylation of Rab11A at serine 177, which inhibits Rab11A and Rab11A-dependent surface trafficking of β1 subunits. The decrease in surface β1 subunits leads to a reduction in BK channel calcium-sensitivity, inhibition of transient BK currents, and vasoconstriction. We describe a unique mechanism by which a vasoconstrictor inhibits BK channels and identify Rab11A serine 177 as a modulator of arterial contractility.
Collapse
Affiliation(s)
- Xue Zhai
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis
| | - M Dennis Leo
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis
| | - Jonathan H Jaggar
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis.
| |
Collapse
|
8
|
del Río-Sancho S, Cros C, Coutaz B, Cuendet M, Kalia YN. Cutaneous iontophoresis of μ-conotoxin CnIIIC—A potent Na V 1.4 antagonist with analgesic, anaesthetic and myorelaxant properties. Int J Pharm 2017; 518:59-65. [DOI: 10.1016/j.ijpharm.2016.12.054] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 01/14/2023]
|
9
|
|
10
|
Bailey TA, Luan H, Tom E, Bielecki TA, Mohapatra B, Ahmad G, George M, Kelly DL, Natarajan A, Raja SM, Band V, Band H. A kinase inhibitor screen reveals protein kinase C-dependent endocytic recycling of ErbB2 in breast cancer cells. J Biol Chem 2014; 289:30443-30458. [PMID: 25225290 DOI: 10.1074/jbc.m114.608992] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ErbB2 overexpression drives oncogenesis in 20-30% cases of breast cancer. Oncogenic potential of ErbB2 is linked to inefficient endocytic traffic into lysosomes and preferential recycling. However, regulation of ErbB2 recycling is incompletely understood. We used a high-content immunofluorescence imaging-based kinase inhibitor screen on SKBR-3 breast cancer cells to identify kinases whose inhibition alters the clearance of cell surface ErbB2 induced by Hsp90 inhibitor 17-AAG. Less ErbB2 clearance was observed with broad-spectrum PKC inhibitor Ro 31-8220. A similar effect was observed with Go 6976, a selective inhibitor of classical Ca(2+)-dependent PKCs (α, β1, βII, and γ). PKC activation by PMA promoted surface ErbB2 clearance but without degradation, and ErbB2 was observed to move into a juxtanuclear compartment where it colocalized with PKC-α and PKC-δ together with the endocytic recycling regulator Arf6. PKC-α knockdown impaired the juxtanuclear localization of ErbB2. ErbB2 transit to the recycling compartment was also impaired upon PKC-δ knockdown. PMA-induced Erk phosphorylation was reduced by ErbB2 inhibitor lapatinib, as well as by knockdown of PKC-δ but not that of PKC-α. Our results suggest that activation of PKC-α and -δ mediates a novel positive feedback loop by promoting ErbB2 entry into the endocytic recycling compartment, consistent with reported positive roles for these PKCs in ErbB2-mediated tumorigenesis. As the endocytic recycling compartment/pericentrion has emerged as a PKC-dependent signaling hub for G-protein-coupled receptors, our findings raise the possibility that oncogenesis by ErbB2 involves previously unexplored PKC-dependent endosomal signaling.
Collapse
Affiliation(s)
- Tameka A Bailey
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950
| | - Haitao Luan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950; Departments of Genetics, Cell Biology, and Anatomy, and University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950
| | - Eric Tom
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950; Departments of Biochemistry & Molecular Biology, College of Medicine, and University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950
| | - Timothy Alan Bielecki
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950
| | - Bhopal Mohapatra
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950; Departments of Biochemistry & Molecular Biology, College of Medicine, and University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950
| | - Gulzar Ahmad
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950
| | - Manju George
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950
| | - David L Kelly
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950
| | - Amarnath Natarajan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950
| | - Srikumar M Raja
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950
| | - Vimla Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950; Departments of Genetics, Cell Biology, and Anatomy, and University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950
| | - Hamid Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950; Departments of Genetics, Cell Biology, and Anatomy, and University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950; Departments of Biochemistry & Molecular Biology, College of Medicine, and University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, Nebraska 68198-5950.
| |
Collapse
|
11
|
Mitogenic effects of phosphatidylcholine nanoparticles on MCF-7 breast cancer cells. BIOMED RESEARCH INTERNATIONAL 2014; 2014:687037. [PMID: 24772432 PMCID: PMC3977480 DOI: 10.1155/2014/687037] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/14/2014] [Accepted: 02/14/2014] [Indexed: 02/06/2023]
Abstract
Lecithins, mainly composed of the phospholipids phosphatidylcholines (PC), have many different uses in the pharmaceutical and clinical field. PC are involved in structural and biological functions as membrane trafficking processes and cellular signaling. Considering the increasing applications of lecithin-based nanosystems for the delivery of therapeutic agents, the aim of the present work was to determine the effects of phosphatidylcholine nanoparticles over breast cancer cellular proliferation and signaling. PC dispersions at 0.01 and 0.1% (w/v) prepared in buffer pH 7.0 and 5.0 were studied in the MCF-7 breast cancer cell line. Neutral 0.1% PC-derived nanoparticles induced the activation of the MEK-ERK1/2 pathway, increased cell viability and induced a 1.2 fold raise in proliferation. These biological effects correlated with the increase of epidermal growth factor receptor (EGFR) content and its altered cellular localization. Results suggest that nanoparticles derived from PC dispersion prepared in buffer pH 7.0 may induce physicochemical changes in the plasma membrane of cancer cells which may affect EGFR cellular localization and/or activity, increasing activation of the MEK-ERK1/2 pathway and inducing proliferation. Results from the present study suggest that possible biological effects of delivery systems based on lecithin nanoparticles should be taken into account in pharmaceutical formulation design.
Collapse
|
12
|
Liu M, Idkowiak-Baldys J, Roddy PL, Baldys A, Raymond J, Clarke CJ, Hannun YA. Sustained activation of protein kinase C induces delayed phosphorylation and regulates the fate of epidermal growth factor receptor. PLoS One 2013; 8:e80721. [PMID: 24244711 PMCID: PMC3823608 DOI: 10.1371/journal.pone.0080721] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 10/04/2013] [Indexed: 01/11/2023] Open
Abstract
It is well established that acute activation of members of the protein kinase C (PKC) family induced by activation of cellular receptors can transduce extracellular stimuli to intracellular signaling. However, the functions of sustained activation of PKC are not well studied. We have previously shown that sustained activation of classical PKC isoforms over 15-60 min induced the formation of the pericentrion, a subset of recycling endosomes that are sequestered perinuclearly in a PKC- and phospholipase D (PLD)-dependent manner. In this study, we investigated the role of this process in the phosphorylation of EGFR on threonine 654 (Thr-654) and in the regulation of intracellular trafficking and fate of epidermal growth factor receptor (EGFR). Sustained stimulation of the angiotensin II receptor induced translocation of the EGFR to the pericentrion, which in turn prevents full access of EGF to the EGFR. These effects required PKC and PLD activities, and direct stimulation of PKC with phorbol esters was sufficient to reproduce these effects. Furthermore, activation of PKC induced delayed phosphorylation of EGFR on Thr-654 that coincided with the formation of the pericentrion and which was dependent on PLD and endocytosis of EGFR. Thus, Thr-654 phosphorylation required the formation of the pericentrion. On the other hand, using a T654A mutant of EGFR, we find that the phosphorylation on Thr-654 was not required for translocation of EGFR to the pericentrion but was required for protection of EGFR from degradation in response to EGF. Taken together, these results demonstrate a novel role for the pericentrion in the regulation of EGFR phosphorylation, which in turn is important for the fates of EGFR.
Collapse
Affiliation(s)
- Mengling Liu
- Department of Medicine and The Stony Brook Cancer Center, Stony Brook University, Stony Brook, New York, United States of America
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Jolanta Idkowiak-Baldys
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Patrick L. Roddy
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Aleksander Baldys
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Medical and Research Services, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, United States of America
| | - John Raymond
- Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Christopher J. Clarke
- Department of Medicine and The Stony Brook Cancer Center, Stony Brook University, Stony Brook, New York, United States of America
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Yusuf A. Hannun
- Department of Medicine and The Stony Brook Cancer Center, Stony Brook University, Stony Brook, New York, United States of America
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
13
|
El Osta M, Liu M, Adada M, Senkal CE, Idkowiak-Baldys J, Obeid LM, Clarke CJ, Hannun YA. Sustained PKCβII activity confers oncogenic properties in a phospholipase D- and mTOR-dependent manner. FASEB J 2013; 28:495-505. [PMID: 24121461 DOI: 10.1096/fj.13-230557] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Protein kinase C (PKC) is a family of serine/threonine kinases implicated in a variety of physiological processes. We have shown previously that sustained activation of the classical PKCα and PKCβII induces their phospholipase D (PLD)-dependent internalization and translocation to a subset of the recycling endosomes defined by the presence of PKC and PLD (the pericentrion), which results in significant differences in phosphorylation of PKC substrates. Here, we have investigated the biological consequences of sustained PKC activity and the involvement of PLD in this process. We find that sustained activation of PKC results in activation of the mammalian target of rapamycin (mTOR)/S6 kinase pathway in a PLD- and endocytosis-dependent manner, with both pharmacologic inhibitors and siRNA implicating the PLD2 isoform. Notably, dysregulated overexpression of PKCβII in A549 lung cancer cells was necessary for the enhanced proliferation and migration of these cancer cells. Inhibition of PKCβII with enzastaurin reduced A549 cell proliferation by >60% (48 h) and migration by >50%. These biological effects also required both PLD activity and mTOR function, with both the PLD inhibitor FIPI and rapamycin reducing cell growth by >50%. Reciprocally, forced overexpression of wild-type PKCβII, but not an F666D mutant that cannot interact with PLD, was sufficient to enhance cell growth and increase migration of noncancerous HEK cells; indeed, both properties were almost doubled when compared to vector control and PKC-F666D-overexpressing cells. Notably, this condition was also dependent on both PLD and mTOR activity. In summary, these data define a PKC-driven oncogenic signaling pathway that requires both PLD and mTOR, and suggest that inhibitors of PLD or mTOR would be beneficial in cancers where PKC overexpression is a contributing or driving factor.
Collapse
Affiliation(s)
- Mohamad El Osta
- 2Stony Brook Cancer Center and Department of Medicine, Stony Brook University, 100 Nicolls Rd., Stony Brook, NY 11794, USA.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Er EE, Mendoza MC, Mackey AM, Rameh LE, Blenis J. AKT facilitates EGFR trafficking and degradation by phosphorylating and activating PIKfyve. Sci Signal 2013; 6:ra45. [PMID: 23757022 DOI: 10.1126/scisignal.2004015] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase (RTK) that controls cell proliferation, growth, survival, metabolism, and migration by activating the PI3K (phosphatidylinositol 3-kinase)-AKT and ERK (extracellular signal-regulated kinase)-RSK (ribosomal S6 kinase) pathways. EGFR signaling to these pathways is temporally and spatially regulated. Endocytic trafficking controls the access of EGFR to these downstream effectors and also its degradation, which terminates EGFR signaling. We showed that AKT facilitated the endocytic trafficking of EGFR to promote its degradation. Interfering with AKT signaling reduced both EGFR recycling and the rate of EGFR degradation. In AKT-impaired cells, EGFRs were unable to reach the cell surface or the lysosomal compartment and accumulated in the early endosomes, resulting in prolonged signaling and increased activation of ERK and RSK. Upon EGF stimulation, AKT phosphorylated and activated the kinase PIKfyve [FYVE-containing phosphatidylinositol 3-phosphate 5-kinase], which promoted vesicle trafficking to lysosomes. PIKfyve activation promoted EGFR degradation. Similar regulation occurred with platelet-derived growth factor receptor (PDGFR), suggesting that AKT phosphorylation and activation of PIKfyve is likely to be a common feedback mechanism for terminating RTK signaling and reducing receptor abundance.
Collapse
Affiliation(s)
- Ekrem Emrah Er
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
15
|
The role of endosomal signaling triggered by metastatic growth factors in tumor progression. Cell Signal 2013; 25:1539-45. [PMID: 23571269 DOI: 10.1016/j.cellsig.2013.03.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Accepted: 03/28/2013] [Indexed: 01/12/2023]
Abstract
Within tumor microenvironment, a lot of growth factors such as hepatocyte growth factor and epidermal growth factor may induce similar signal cascade downstream of receptor tyrosine kinase (RTK) and trigger tumor metastasis synergistically. In the past decades, the intimate relationship of RTK-mediated receptor endocytosis with signal transduction was well established. In general, most RTK undergoes clathrin-dependent endocytosis and/or clathrin-independent endocytosis. The internalized receptors may sustain the signaling within early endosome, recycling to plasma membrane for subsequent ligand engagement or sorting to late endosomes/lysosome for receptor degradation. Moreover, receptor endocytosis influences signal transduction in a temporal and spatial manner for periodical and polarized cellular processes such as cell migration. The endosomal signalings triggered by various metastatic factors are quite similar in some critical points, which are essential for triggering cell migration and tumor progression. There are common regulators for receptor endocytosis including dynamin, Rab4, Rab5, Rab11 and Cbl. Moreover, many critical regulators within the RTK signal pathway such as Grb2, p38, PKC and Src were also modulators of endocytosis. In the future, these may constitute a new category of targets for prevention of tumor metastasis.
Collapse
|
16
|
Thompson PM, Cruz DA, Olukotun DY, Delgado PL. Serotonin receptor, SERT mRNA and correlations with symptoms in males with alcohol dependence and suicide. Acta Psychiatr Scand 2012; 126:165-74. [PMID: 22176604 DOI: 10.1111/j.1600-0447.2011.01816.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE This study tested the hypothesis that abnormalities in components of the serotonin (5HT) system in the prefrontal cortex are associated with suicide in alcohol-dependent subjects. Second, we assessed the relationship of lifetime impulsivity and mood symptoms with prefrontal cortex 5-HT measures. METHOD Tissue was obtained from Brodmann's areas (BA) 9 and 24 in postmortem samples of individuals who were alcohol dependent with suicide (n = 5), alcohol dependent without suicide (n = 9) and normal controls (n = 5). Serotonin receptor (5HT) and serotonin reuptake transporter (SERT) mRNA were measured. Interviews with next of kin estimated lifetime impulsivity and mood symptoms in the last week of life. RESULTS Serotonin receptor 1A (5HT1A) mRNA in BA 9 was elevated in the alcohol dependence without suicide group compared with controls. In the alcohol dependence with suicide group, anxiety symptoms were associated with decreased BA 24 SERT mRNA and depressive symptoms with BA 9 5HT1A mRNA expression. In the alcohol dependent only group impulsivity is correlated with increased BA 9, and BA 24 serotonin receptor 2A mRNA. CONCLUSION Our data suggest region-specific change, rather than global serotonin blunting is involved in alcohol dependence and suicide. It also suggests that symptoms are differentially influenced by prefrontal cortex serotonin receptor mRNA levels.
Collapse
Affiliation(s)
- P M Thompson
- Department of Psychiatry, University of Texas Health Science Center San Antonio Southwest Brain Bank, USA.
| | | | | | | |
Collapse
|
17
|
Kolesnikov YS, Nokhrina KP, Kretynin SV, Volotovski ID, Martinec J, Romanov GA, Kravets VS. Molecular structure of phospholipase D and regulatory mechanisms of its activity in plant and animal cells. BIOCHEMISTRY (MOSCOW) 2012; 77:1-14. [DOI: 10.1134/s0006297912010014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Pavarotti M, Capmany A, Vitale N, Colombo MI, Damiani MT. Rab11 is phosphorylated by classical and novel protein kinase C isoenzymes upon sustained phorbol ester activation. Biol Cell 2012; 104:102-15. [PMID: 22188018 DOI: 10.1111/boc.201100062] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Accepted: 11/29/2011] [Indexed: 01/06/2023]
Abstract
BACKGROUND INFORMATION Rab11 is a small GTPase that controls diverse intracellular trafficking pathways. However, the molecular machinery that regulates the participation of Rab11 in those different transport events is poorly understood. In resting cells, Rab11 localizes at the endocytic recycling compartment (ERC), whereas the different protein kinase C (PKC) isoforms display a cytosolic distribution. RESULTS Sustained phorbol ester stimulation induces the translocation of the classical PKCα and PKCβII isoenzymes to the ERC enriched in Rab11, and results in transferrin recycling inhibition. In contrast, novel PKCε and atypical PKCζ isoenzymes neither redistribute to the perinucleus nor modify transferrin recycling transport after phorbol ester stimulation. Although several Rabs have been shown to be phosphorylated, there is to date no evidence indicating Rab11 as a kinase substrate. In this report, we show that Rab11 appears phosphorylated in vivo in phorbol ester-stimulated cells. A bioinformatic analysis of Rab11 allowed us to identify several high-probability Ser/Thr kinase phosphorylation sites. Our results demonstrate that classical PKC (PKCα and PKCβII but not PKCβI) directly phosphorylate Rab11 in vitro. In addition, novel PKCε and PKCη but not PKCδ isoenzymes also phosphorylate Rab11. Mass spectrometry analysis revealed that Ser 177 is the Rab11 residue to be phosphorylated in vitro by either PKCβII or PKCε. In agreement, the phosphomimetic mutant, Rab11 S177D, retains transferrin at the ERC in the absence of phorbol-12-myristate-13-acetate stimulus. CONCLUSIONS This report shows for the first time that Rab11 is differentially phosphorylated by distinct PKC isoenzymes and that this post-translational modification might be a regulatory mechanism of intracellular trafficking.
Collapse
Affiliation(s)
- Martín Pavarotti
- IHEM-CONICET, National Research Council, School of Medicine, University of Cuyo, Mendoza, Argentina
| | | | | | | | | |
Collapse
|
19
|
Li X, DiFiglia M. The recycling endosome and its role in neurological disorders. Prog Neurobiol 2011; 97:127-41. [PMID: 22037413 DOI: 10.1016/j.pneurobio.2011.10.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 10/14/2011] [Accepted: 10/17/2011] [Indexed: 02/08/2023]
Abstract
The recycling endosome (RE) is an organelle in the endocytic pathway where plasma membranes (proteins and lipids) internalized by endocytosis are processed back to the cell surface for reuse. Endocytic recycling is the primary way for the cell to maintain constituents of the plasma membrane (Griffiths et al., 1989), i.e., to maintain the abundance of receptors and transporters on cell surfaces. Membrane traffic through the RE is crucial for several key cellular processes including cytokinesis and cell migration. In polarized cells, including neurons, the RE is vital for the generation and maintenance of the polarity of the plasma membrane. Many RE dependent cargo molecules are known to be important for neuronal function and there is evidence that improper function of key proteins in RE-associated pathways may contribute to the pathogenesis of neurological disorders, including Huntington's disease. The function of the RE in neurons is poorly understood. Therefore, there is need to understand how membrane dynamics in RE-associated pathways are affected or participate in the development or progression of neurological diseases. This review summarizes advances in understanding endocytic recycling associated with the RE, challenges in elucidating molecular mechanisms underlying RE function, and evidence for RE dysfunction in neurological disorders.
Collapse
Affiliation(s)
- Xueyi Li
- Laboratory of Cellular Neurobiology and Department of Neurology, Massachusetts General Hospital, 114 16th Street, Charlestown, MA 02129, USA
| | | |
Collapse
|
20
|
El-Shewy HM, Abdel-Samie SA, Al Qalam AM, Lee MH, Kitatani K, Anelli V, Jaffa AA, Obeid LM, Luttrell LM. Phospholipase C and protein kinase C-β 2 mediate insulin-like growth factor II-dependent sphingosine kinase 1 activation. Mol Endocrinol 2011; 25:2144-56. [PMID: 22016563 DOI: 10.1210/me.2011-0101] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
We recently reported that IGF-II binding to the IGF-II/mannose-6-phosphate (M6P) receptor activates the ERK1/2 cascade by triggering sphingosine kinase 1 (SK1)-dependent transactivation of G protein-coupled sphingosine 1-phosphate (S1P) receptors. Here, we investigated the mechanism of IGF-II/M6P receptor-dependent sphingosine kinase 1 (SK1) activation in human embryonic kidney 293 cells. Pretreating cells with protein kinase C (PKC) inhibitor, bisindolylmaleimide-I, abolished IGF-II-stimulated translocation of green fluorescent protein (GFP)-tagged SK1 to the plasma membrane and activation of endogenous SK1, implicating PKC as an upstream regulator of SK1. Using confocal microscopy to examine membrane translocation of GFP-tagged PKCα, β1, β2, δ, and ζ, we found that IGF-II induced rapid, transient, and isoform-specific translocation of GFP-PKCβ2 to the plasma membrane. Immunoblotting of endogenous PKC phosphorylation confirmed PKCβ2 activation in response to IGF-II. Similarly, IGF-II stimulation caused persistent membrane translocation of the kinase-deficient GFP-PKCβ2 (K371R) mutant, which does not dissociate from the membrane after translocation. IGF-II stimulation increased diacylglycerol (DAG) levels, the established activator of classical PKC. Interestingly, the polyunsaturated fraction of DAG was increased, indicating involvement of phosphatidyl inositol/phospholipase C (PLC). Pretreating cells with the PLC inhibitor, U73122, attenuated IGF-II-dependent DAG production and PKCβ2 phosphorylation, blocked membrane translocation of the kinase-deficient GFP-PKCβ2 (K371R) mutant, and reduced sphingosine 1-phosphate production, suggesting that PLC/PKCβ2 are upstream regulators of SK1 in the pathway. Taken together, these data provide evidence that activation of PLC and PKCβ2 by the IGF-II/M6P receptor are required for the activation of SK1.
Collapse
Affiliation(s)
- Hesham M El-Shewy
- Departments of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
El-Osta MA, Idkowiak-Baldys J, Hannun YA. Delayed phosphorylation of classical protein kinase C (PKC) substrates requires PKC internalization and formation of the pericentrion in a phospholipase D (PLD)-dependent manner. J Biol Chem 2011; 286:19340-53. [PMID: 21478146 DOI: 10.1074/jbc.m110.152330] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
It was previously demonstrated that sustained activation (30-60 min) of protein kinase C (PKC) results in translocation of PKC α and βII to the pericentrion, a dynamic subset of the recycling compartment whose formation is dependent on PKC and phospholipase D (PLD). Here we investigated whether the formation of the pericentrion modulates the ability of PKC to phosphorylate substrates, especially if it reduces substrate phosphorylation by sequestering PKC. Surprisingly, using an antibody that detects phosphosubstrates of classical PKCs, the results showed that the majority of PKC phosphosubstrates are phosphorylated with delayed kinetics, correlating with the time frame of PKC translocation to the pericentrion. Substrate phosphorylation was blocked by PLD inhibitors and was not observed in response to activation of a PKC βII mutant (F663D) that is defective in interaction with PLD and in internalization. Phosphorylation was also inhibited by blocking clathrin-dependent endocytosis, demonstrating a requirement for endocytosis for the PKC-dependent major phosphorylation effects. Serotonin receptor activation by serotonin showed a similar response to phorbol 12-myristate 13-acetate, implicating a potential role of delayed kinetics in G protein-coupled receptor signaling. Evaluation of candidate substrates revealed that the phosphorylation of the PKC substrate p70S6K kinase behaved in a similar manner. Gradient-based fractionation revealed that the majority of these PKC substrates reside within the pericentrion-enriched fractions and not in the plasma membrane. Finally, proteomic analysis of the pericentrion-enriched fractions revealed several proteins as known PKC substrates and/or proteins involved in endocytic trafficking. These results reveal an important role for PKC internalization and for the pericentrion as key determinants/amplifiers of PKC action.
Collapse
Affiliation(s)
- Mohamad A El-Osta
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | |
Collapse
|
22
|
Baldys A, Raymond JR. Role of c-Cbl carboxyl terminus in serotonin 5-HT2A receptor recycling and resensitization. J Biol Chem 2011; 286:24656-65. [PMID: 21464131 DOI: 10.1074/jbc.m110.119891] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The 5-hydroxytryptamine 2A receptor (5-HT(2A)R) undergoes constitutive and agonist-dependent internalization. Despite many advances in our understanding of G protein-coupled receptor trafficking, the exact mechanism of endocytic sorting of G protein-coupled receptors remains obscure. Recently, we have reported a novel finding documenting a global role for the ubiquitin ligase c-Cbl in regulating vesicular sorting of epidermal growth factor receptor (Baldys, A., Göoz, M., Morinelli, T. A., Lee, M. H., Raymond, J. R., Jr., Luttrell, L. M., and Raymond, J. R., Sr. (2009) Biochemistry 48, 1462-1473). Thus, we tested the hypothesis that c-Cbl might play a role in 5-HT(2A)R recycling. In this study, we demonstrated an association of 5-HT(2A)R with c-Cbl. Furthermore, down-regulation of c-Cbl by RNA interference blocked efficient recycling of 5-HT(2A)R to the plasma membrane. Immunofluorescence microscopy revealed that 5-HT(2A) receptors were trapped in early endosome antigen 1- and Rab11-positive sorting endosomes in cells overexpressing c-Cbl mutants lacking carboxyl termini. This inhibitory effect was associated with a relative decrease in association of c-Cbl truncation proteins with the 5-HT(2A)R, compared with that observed for the full-length c-Cbl fusion protein. Consistent with the delayed recycling, 5-HT(2A)R resensitization was greatly attenuated in the presence of c-Cbl mutants lacking carboxyl termini, as detected by changes in the cytosolic calcium. Taken together, these studies have led to the discovery that the C-terminal region of c-Cbl plays a crucial role in the temporal and spatial control of 5-HT(2A)R recycling.
Collapse
Affiliation(s)
- Aleksander Baldys
- Nephrology Division, Department of Medicine, Medical University of South Carolina, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29425, USA
| | | |
Collapse
|
23
|
Zaarour N, Defontaine N, Demaretz S, Azroyan A, Cheval L, Laghmani K. Secretory carrier membrane protein 2 regulates exocytic insertion of NKCC2 into the cell membrane. J Biol Chem 2011; 286:9489-9502. [PMID: 21205824 PMCID: PMC3059028 DOI: 10.1074/jbc.m110.166546] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 12/23/2010] [Indexed: 12/20/2022] Open
Abstract
The renal-specific Na-K-2Cl co-transporter, NKCC2, plays a pivotal role in regulating body salt levels and blood pressure. NKCC2 mutations lead to type I Bartter syndrome, a life-threatening kidney disease. Regulation of NKCC2 trafficking behavior serves as a major mechanism in controlling NKCC2 activity across the plasma membrane. However, the identities of the protein partners involved in cell surface targeting of NKCC2 are largely unknown. To gain insight into these processes, we used a yeast two-hybrid system to screen a kidney cDNA library for proteins that interact with the NKCC2 C terminus. One binding partner we identified was SCAMP2 (secretory carrier membrane protein 2). Microscopic confocal imaging and co-immunoprecipitation assays confirmed NKCC2-SCAMP2 interaction in renal cells. SCAMP2 associated also with the structurally related co-transporter NCC, suggesting that the interaction with SCAMP2 is a common feature of sodium-dependent chloride co-transporters. Heterologous expression of SCAMP2 specifically decreased cell surface abundance as well as transport activity of NKCC2 across the plasma membrane. Co-immunolocalization experiments revealed that intracellularly retained NKCC2 co-localizes with SCAMP2 in recycling endosomes. The rate of NKCC2 endocytic retrieval, assessed by the sodium 2-mercaptoethane sulfonate cleavage assay, was not affected by SCAMP2. The surface-biotinylatable fraction of newly inserted NKCC2 in the plasma membrane was reduced by SCAMP2, demonstrating that SCAMP2-induced decrease in surface NKCC2 is due to decreased exocytotic trafficking. Finally, a single amino acid mutation, cysteine 201 to alanine, within the conserved cytoplasmic E peptide of SCAMP2, which is believed to regulate exocytosis, abolished SCAMP2-mediated down-regulation of the co-transporter. Taken together, these data are consistent with a model whereby SCAMP2 regulates NKCC2 transit through recycling endosomes and limits the cell surface targeting of the co-transporter by interfering with its exocytotic trafficking.
Collapse
Affiliation(s)
- Nancy Zaarour
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| | - Nadia Defontaine
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| | - Sylvie Demaretz
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| | - Anie Azroyan
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| | - Lydie Cheval
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| | - Kamel Laghmani
- From INSERM, Centre de Recherche des Cordeliers, UMRS 872, CNRS, ERL7226, 75006 Paris, France
- Université Pierre et Marie Curie, 75006 Paris, France, and
- the Faculté de Médecine, Université Paris-Descartes, 75005 Paris, France
| |
Collapse
|
24
|
Abstract
Brain serotonergic circuitries interact with other neurotransmitter systems on a multitude of different molecular levels. In humans, as in other mammalian species, serotonin (5-HT) plays a modulatory role in almost every physiological function. Furthermore, serotonergic dysfunction is thought to be implicated in several psychiatric and neurodegenerative disorders. We describe the neuroanatomy and neurochemistry of brain serotonergic circuitries. The contribution of emergent in vivo imaging methods to the regional localization of binding site receptors and certain aspects of their functional connectivity in correlation to behavior is also discussed. 5-HT cell bodies, mainly localized in the raphe nuclei, send axons to almost every brain region. It is argued that the specificity of the local chemocommunication between 5-HT and other neuronal elements mainly depends on mechanisms regulating the extracellular concentration of 5-HT the diversity of high-affinity membrane receptors, and their specific transduction modalities.
Collapse
Affiliation(s)
- Yves Charnay
- Hôpitaux Universitaires de Genève, Département de Psychiatrie, Service de Neuropsychiatrie, Ch. Du Petit-Bel-Air, 2, CH-1225 Chêne-Bourg, Switzerland.
| | | |
Collapse
|
25
|
Rodriguez AR, Yu JJ, Murthy AK, Guentzel MN, Klose KE, Forsthuber TG, Chambers JP, Berton MT, Arulanandam BP. Mast cell/IL-4 control of Francisella tularensis replication and host cell death is associated with increased ATP production and phagosomal acidification. Mucosal Immunol 2011; 4:217-26. [PMID: 20861832 PMCID: PMC3040285 DOI: 10.1038/mi.2010.59] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Mast cells are now recognized as effective modulators of innate immunity. We recently reported that mast cells and secreted interleukin-4 (IL-4) effectively control intramacrophage replication of Francisella tularensis Live Vaccine Strain (LVS), and that mice deficient in mast cells or IL-4 receptor (IL-4R(-/-)) exhibit greater susceptibility to pulmonary challenge. In this study, we further evaluated the mechanism(s) by which mast cells/IL-4 control intramacrophage bacterial replication and host cell death, and found that IL-4R(-/-) mice exhibited significantly greater induction of active caspase-3 within lung macrophages than wild-type animals following intranasal challenge with either LVS or the human virulent type A strain SCHU S4. Treatment of LVS-infected bone-marrow-derived macrophages with a pancaspase inhibitor (zVAD) did not alter bacterial replication, but minimized active caspase-3 and other markers (Annexin V and propidium iodide) of cell death, whereas treatment with both rIL-4 and zVAD resulted in concomitant reduction of both parameters, suggesting that inhibition of bacterial replication by IL-4 was independent of caspase activation. Interestingly, IL-4-treated infected macrophages exhibited significantly increased ATP production and phagolysosomal acidification, as well as enhanced mannose receptor upregulation and increased internalization with acidification, which correlated with observations in mast cell-macrophage co-cultures, with resultant decreases in F. tularensis replication.
Collapse
MESH Headings
- Adenosine Triphosphate/biosynthesis
- Animals
- Caspase 3/metabolism
- Cell Death/immunology
- Cells, Cultured
- Enzyme Activation/drug effects
- Enzyme Inhibitors/pharmacology
- Francisella tularensis/growth & development
- Francisella tularensis/immunology
- Gene Expression Regulation
- Host-Pathogen Interactions
- Interleukin-4/immunology
- Lectins, C-Type/metabolism
- Macrophages, Alveolar/enzymology
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/pathology
- Mannose Receptor
- Mannose-Binding Lectins/metabolism
- Mast Cells/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Oligopeptides/pharmacology
- Organelles/chemistry
- Organelles/microbiology
- Phagosomes/chemistry
- Phagosomes/immunology
- Receptors, Cell Surface/metabolism
- Receptors, Interleukin-4/genetics
- Receptors, Interleukin-4/immunology
- Signal Transduction/immunology
- Tularemia/immunology
Collapse
Affiliation(s)
- Annette R. Rodriguez
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249; USA
| | - Jieh-Juen Yu
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249; USA
| | - Ashlesh K. Murthy
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249; USA
| | - M. Neal Guentzel
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249; USA
| | - Karl E. Klose
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249; USA
| | - Thomas G. Forsthuber
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249; USA
| | - James P. Chambers
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249; USA
| | - Michael T. Berton
- Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio, TX 78229; USA
| | - Bernard. P. Arulanandam
- South Texas Center for Emerging Infectious Diseases, Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249; USA
| |
Collapse
|
26
|
Welling PA, Weisz OA. Sorting it out in endosomes: an emerging concept in renal epithelial cell transport regulation. Physiology (Bethesda) 2011; 25:280-92. [PMID: 20940433 DOI: 10.1152/physiol.00022.2010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Ion and water transport by the kidney is continually adjusted in response to physiological cues. Selective endocytosis and endosomal trafficking of ion transporters are increasingly appreciated as mechanisms to acutely modulate renal function. Here, we discuss emerging paradigms in this new area of investigation.
Collapse
Affiliation(s)
- Paul A Welling
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
27
|
Milhas D, Clarke CJ, Idkowiak-Baldys J, Canals D, Hannun YA. Anterograde and retrograde transport of neutral sphingomyelinase-2 between the Golgi and the plasma membrane. BIOCHIMICA ET BIOPHYSICA ACTA 2010; 1801:1361-74. [PMID: 20713176 PMCID: PMC2957534 DOI: 10.1016/j.bbalip.2010.08.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 07/30/2010] [Accepted: 08/02/2010] [Indexed: 02/06/2023]
Abstract
The activation of neutral sphingomyelinase-2 (nSMase2) and consequent ceramide production are implicated in many stress-induced signaling pathways. Trafficking of nSMase2 from the Golgi compartment to the plasma membrane (PM) in response to signaling stimuli has been described. However, the precise mechanisms of transport remain unknown. This study aimed to investigate the trafficking of nSMase2 between the Golgi and the PM. We show here that V5-nSMase2 localizes at the PM and Golgi in MCF-7 cells and confirm relocalization of nSMase2 to the PM at confluence. Although cycloheximide (CHX) treatment partially inhibited the Golgi localization of GFP-nSMase2, recovery of GFP-nSMase2 to an intracellular compartment was still observed after photobleaching. Moreover, in the presence of CHX, GFP- and V5-nSMase2 co-localized with endosomal/recycling markers. In HEK293 cells, activation of either protein kinase C-alpha or betaII, with the phorbol ester PMA led to relocalization of both wild-type and inactive nSMase2 to the pericentrion, a PKC-dependent subset of recycling endosomes. Finally, inhibition of nSMase2 endocytosis by K+depletion reduced the intracellular pool of nSMase2 and increased nSMase2 activity resulting in elevated ceramide levels. Altogether, these results suggest that nSMase2 traffics from the Golgi to the PM as a membrane protein en route to the cell surface and recycles back to the Golgi through the endosomal/recycling compartment. Moreover, the recycling of nSMase2 from the PM is important for its catalytic regulation.
Collapse
Affiliation(s)
- Delphine Milhas
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | - Daniel Canals
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Yusuf A. Hannun
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
28
|
Bocock JP, Carmicle S, Sircar M, Erickson AH. Trafficking and proteolytic processing of RNF13, a model PA-TM-RING family endosomal membrane ubiquitin ligase. FEBS J 2010; 278:69-77. [PMID: 21078126 DOI: 10.1111/j.1742-4658.2010.07924.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
RING finger protein 13 (RNF13) is a ubiquitously expressed, highly regulated ubiquitin ligase anchored in endosome membranes. A RING domain located in the cytoplasmic half of this type 1 membrane protein mediates ubiquitination in vitro but physiological substrates have not yet been identified. The protein localized in endosomal membranes undergoes extensive proteolysis in a proteasome-dependent manner, but the mRNA level can be increased and the encoded protein stabilized under specific physiological conditions. The cytoplasmic half of RNF13 is released from the membrane by regulatory proteases and therefore has the potential to mediate ubiquitination at distant sites independent of the full-length protein. In response to protein kinase C activation, the full-length protein is stabilized and moves to recycling endosomes and to the inner nuclear membrane, which exposes the RING domain to the nucleoplasm. Thus RNF13 is a ubiquitin ligase that can potentially mediate ubiquitination in endosomes, on the plasma membrane, in the cytoplasm, in the nucleoplasm or on the inner nuclear membrane, with the site(s) regulated by signaling events that modulate protein targeting and proteolysis.
Collapse
Affiliation(s)
- Jeffrey P Bocock
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | |
Collapse
|
29
|
Cai B, Katafiasz D, Horejsi V, Naslavsky N. Pre-sorting endosomal transport of the GPI-anchored protein, CD59, is regulated by EHD1. Traffic 2010; 12:102-20. [PMID: 20961375 DOI: 10.1111/j.1600-0854.2010.01135.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
EHD1 regulates the trafficking of multiple receptors from the endocytic recycling compartment (ERC) to the plasma membrane. However, the potential role of EHD1 in regulating the family of glycosylphosphatidylinositol-anchored proteins (GPI-APs) has not been determined. Here we demonstrate a novel role for EHD1 in regulating the trafficking of CD59, an endogenous GPI-AP, at early stages of trafficking through the endocytic pathway. EHD1 displays significant colocalization with newly internalized CD59. Upon EHD1 depletion, there is a rapid Rab5-independent coalescence of CD59 in the ERC region. However, expression of an active Arf6 mutant (Q67L), which traps internalized pre-sorting endosomal cargo in phosphatidylinositol(4,5)-bisphosphate enriched vacuoles, prevents this coalescence. It is of interest that sustained PKC activation leads to a similar coalescence of CD59 at the ERC, and treatment of EHD1-depleted cells with a PKC inhibitor (Go6976) blocked this rapid relocation of CD59. However, unlike sustained PKC activation, EHD1 depletion does not induce the translocation of PKCα to ERC. The results presented herein provide evidence that EHD1 is involved in the control of CD59 transport from pre-sorting endosomes to the ERC in a PKC-dependent manner. However, the mechanisms of EHD1-induced coalescence of CD59 at the ERC differ from those induced by sustained PKC activation.
Collapse
Affiliation(s)
- Bishuang Cai
- Department of Biochemistry and Molecular Biology and Eppley Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | | | | | | |
Collapse
|
30
|
Jenkins RW, Canals D, Idkowiak-Baldys J, Simbari F, Roddy P, Perry DM, Kitatani K, Luberto C, Hannun YA. Regulated secretion of acid sphingomyelinase: implications for selectivity of ceramide formation. J Biol Chem 2010; 285:35706-18. [PMID: 20807762 DOI: 10.1074/jbc.m110.125609] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The acid sphingomyelinase (aSMase) gene gives rise to two distinct enzymes, lysosomal sphingomyelinase (L-SMase) and secretory sphingomyelinase (S-SMase), via differential trafficking of a common protein precursor. However, the regulation of S-SMase and its role in cytokine-induced ceramide formation remain ill defined. To determine the role of S-SMase in cellular sphingolipid metabolism, MCF7 breast carcinoma cells stably transfected with V5-aSMase(WT) were treated with inflammatory cytokines. Interleukin-1β and tumor necrosis factor-α induced a time- and dose-dependent increase in S-SMase secretion and activity, coincident with selective elevations in cellular C(16)-ceramide. To establish a role for S-SMase, we utilized a mutant of aSMase (S508A) that is shown to retain L-SMase activity, but is defective in secretion. MCF7 expressing V5-aSMase(WT) exhibited increased S-SMase and L-SMase activity, as well as elevated cellular levels of specific long-chain and very long-chain ceramide species relative to vector control MCF7. Interestingly, elevated levels of only certain very long-chain ceramides were evident in V5-aSMase(S508A) MCF7. Secretion of the S508A mutant was also defective in response to IL-1β, as was the regulated generation of C(16)-ceramide. Taken together, these data support a crucial role for Ser(508) in the regulation of S-SMase secretion, and they suggest distinct metabolic roles for S-SMase and L-SMase.
Collapse
Affiliation(s)
- Russell W Jenkins
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Crisp KM, Grupe RE, Lobsang TT, Yang X. Biogenic amines modulate pulse rate in the dorsal blood vessel of Lumbriculus variegatus. Comp Biochem Physiol C Toxicol Pharmacol 2010; 151:467-72. [PMID: 20167287 DOI: 10.1016/j.cbpc.2010.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Revised: 02/03/2010] [Accepted: 02/03/2010] [Indexed: 11/28/2022]
Abstract
The biogenic amines are widespread regulators of physiological processes, and play an important role in regulating heart rate in diverse organisms. Here, we present the first pharmacological evidence for a role of the biogenic amines in the regulation of dorsal blood vessel pulse rate in an aquatic oligochaete, Lumbriculus variegatus (Müller, 1774). Bath application of octopamine to intact worms resulted in an acceleration of pulse rate, but not when co-applied with the adenylyl cyclase inhibitor MDL-12,330a. The phosphodiesterase inhibitor theophylline mimicked the effects of OA, but the polar adenosine receptor antagonist 8(p-sulphophenyl)theophylline was significantly less potent than theophylline. Pharmacologically blocking synaptic reuptake of the biogenic amines using the selective 5-HT reuptake blocker fluoxetine or various tricyclic antidepressants also accelerated heart rate. Depletion of the biogenic amines by treatment with the monoamine vesicular transporter blocker reserpine dramatically depressed pulse rate. Pulse rate was partially restored in amine-depleted worms after treatment with octopamine or dopamine, but fully restored following treatment with serotonin. This effect of 5-HT was weakly mimicked by 5-methoxytryptamine, but not by alpha-methylserotonin; it was completely blocked by clozapine and partially blocked by cyproheptadine. Because they are known to orchestrate a variety of adaptive behaviors in invertebrates, the biogenic amines may coordinate blood flow with behavioral state in L.variegatus.
Collapse
Affiliation(s)
- Kevin M Crisp
- Biology Department and Neuroscience Program, St. Olaf College, Northfield, MN 55057, USA.
| | | | | | | |
Collapse
|
32
|
Bocock JP, Carmicle S, Madamba E, Erickson AH. Nuclear targeting of an endosomal E3 ubiquitin ligase. Traffic 2010; 11:756-66. [PMID: 20230530 DOI: 10.1111/j.1600-0854.2010.01060.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Ring finger protein 13 (RNF13) is an E3 ubiquitin ligase embedded in endosome membranes. The protein undergoes constitutive post-translational proteolysis, making its detection difficult unless cells are incubated with a proteasome inhibitor to allow biosynthetic forms to accumulate. When cells were treated with phorbol 12-myristate 13-acetate (PMA), RNF13 avoided proteolysis. A similar stabilization was seen on ionomycin treatment of cells. Drug treatment stabilized both the full-length protein and a membrane-embedded C-terminal fragment generated following ectodomain shedding. Immunofluorescence staining revealed that PMA treatment caused the protein to accumulate in recycling endosomes, where it colocalized with transferrin receptor, and on the inner nuclear membrane, where it colocalized with lamin B. Expression of dominant-negative Rab11 inhibited nuclear localization, suggesting RNF13 was targeted to the inner nuclear membrane through recycling endosomes. New protein synthesis was necessary for this targeting. Nuclear localization was confirmed by immunoelectron microscopy and by purification of the inner nuclear membrane. Stress-induced transport of an endosomal protein to the inner nuclear membrane is a novel mechanism for introduction of regulatory proteins to the DNA environment. RNF13, with its ubiquitin ligase-active RING domain, has the potential to turn over key nuclear proteins in response to signals received at the plasma membrane.
Collapse
Affiliation(s)
- Jeffrey P Bocock
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | |
Collapse
|
33
|
Charnay Y, Léger L. Brain serotonergic circuitries. DIALOGUES IN CLINICAL NEUROSCIENCE 2010; 12:471-87. [PMID: 21319493 PMCID: PMC3181988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2023]
Abstract
Brain serotonergic circuitries interact with other neurotransmitter systems on a multitude of different molecular levels. In humans, as in other mammalian species, serotonin (5-HT) plays a modulatory role in almost every physiological function. Furthermore, serotonergic dysfunction is thought to be implicated in several psychiatric and neurodegenerative disorders. We describe the neuroanatomy and neurochemistry of brain serotonergic circuitries. The contribution of emergent in vivo imaging methods to the regional localization of binding site receptors and certain aspects of their functional connectivity in correlation to behavior is also discussed. 5-HT cell bodies, mainly localized in the raphe nuclei, send axons to almost every brain region. It is argued that the specificity of the local chemocommunication between 5-HT and other neuronal elements mainly depends on mechanisms regulating the extracellular concentration of 5-HT the diversity of high-affinity membrane receptors, and their specific transduction modalities.
Collapse
Affiliation(s)
- Yves Charnay
- Hôpitaux Universitaires de Genève, Département de Psychiatrie, Service de Neuropsychiatrie, Ch. Du Petit-Bel-Air, 2, CH-1225 Chêne-Bourg, Switzerland.
| | | |
Collapse
|