1
|
Johnson KM, Marley MG, Drizyte-Miller K, Chen J, Cao H, Mostafa N, Schott MB, McNiven MA, Razidlo GL. Rab32 regulates Golgi structure and cell migration through Protein Kinase A-mediated phosphorylation of Optineurin. Proc Natl Acad Sci U S A 2025; 122:e2502971122. [PMID: 40258145 PMCID: PMC12054839 DOI: 10.1073/pnas.2502971122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 03/17/2025] [Indexed: 04/23/2025] Open
Abstract
Rab32 is a small GTPase and molecular switch implicated in vesicular trafficking. Rab32 is also an A-Kinase Anchoring Protein (AKAP), which anchors cAMP-dependent Protein Kinase (PKA) to specific subcellular locations and specifies PKA phosphorylation of nearby substrates. Surprisingly, we found that a form of Rab32 deficient in PKA binding (Rab32 L188P) relocalized away from the Golgi apparatus and induced a marked disruption in Golgi organization, assembly, and dynamics. Although Rab32 L188P did not cause a global defect in PKA activity, our data indicate that Rab32 facilitates the phosphorylation of a specific PKA substrate. We uncovered a direct interaction between Rab32 and the adaptor protein optineurin (OPTN), which regulates Golgi dynamics. Further, our data indicate that optineurin is phosphorylated by PKA at Ser342 in a Rab32-dependent manner. Critically, blocking phosphorylation at OPTN Ser342 leads to Golgi fragmentation, and a phospho-mimetic version of OPTN rescues Golgi defects induced by Rab32 L188P. Finally, Rab32 AKAP function and OPTN phosphorylation are required for Golgi repositioning during cell migration, contributing to tumor cell invasion. Together, these data reveal a role for Rab32 in regulating Golgi dynamics through PKA-mediated phosphorylation of OPTN.
Collapse
Affiliation(s)
- Katherine M. Johnson
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN55905
| | - Maxwell G. Marley
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN55905
| | | | - Jing Chen
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN55905
| | - Hong Cao
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN55905
| | - Nourhan Mostafa
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE68198
| | - Micah B. Schott
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN55905
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE68198
| | - Mark A. McNiven
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN55905
| | - Gina L. Razidlo
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN55905
| |
Collapse
|
2
|
Gallo R, Rai AK, McIntyre ABR, Meyer K, Pelkmans L. DYRK3 enables secretory trafficking by maintaining the liquid-like state of ER exit sites. Dev Cell 2023; 58:1880-1897.e11. [PMID: 37643612 DOI: 10.1016/j.devcel.2023.08.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 02/16/2023] [Accepted: 08/01/2023] [Indexed: 08/31/2023]
Abstract
The dual-specificity kinase DYRK3 controls the formation and dissolution of multiple biomolecular condensates, regulating processes including stress recovery and mitotic progression. Here, we report that DYRK3 functionally interacts with proteins associated with endoplasmic reticulum (ER) exit sites (ERESs) and that inhibition of DYRK3 perturbs the organization of the ERES-Golgi interface and secretory trafficking. DYRK3-mediated regulation of ERES depends on the N-terminal intrinsically disordered region (IDR) of the peripheral membrane protein SEC16A, which co-phase separates with ERES components to form liquid-like condensates on the surface of the ER. By modulating the liquid-like properties of ERES, we show that their physical state is essential for functional cargo trafficking through the early secretory pathway. Our findings support a mechanism whereby phosphorylation by DYRK3 and its reversal by serine-threonine phosphatases regulate the material properties of ERES to create a favorable physicochemical environment for directional membrane traffic in eukaryotic cells.
Collapse
Affiliation(s)
- Raffaella Gallo
- Department of Molecular Life Sciences, University of Zurich, 8046 Zurich, Switzerland
| | - Arpan Kumar Rai
- Department of Molecular Life Sciences, University of Zurich, 8046 Zurich, Switzerland.
| | - Alexa B R McIntyre
- Department of Molecular Life Sciences, University of Zurich, 8046 Zurich, Switzerland
| | - Katrina Meyer
- Department of Molecular Life Sciences, University of Zurich, 8046 Zurich, Switzerland
| | - Lucas Pelkmans
- Department of Molecular Life Sciences, University of Zurich, 8046 Zurich, Switzerland.
| |
Collapse
|
3
|
Jia J, Tang S, Yue X, Jing S, Zhu L, Tan C, Gao J, Du Y, Lee I, Qian Y. An A-Kinase Anchoring Protein (ACBD3) Coordinates Traffic-Induced PKA Activation At The Golgi. J Biol Chem 2023; 299:104696. [PMID: 37044218 DOI: 10.1016/j.jbc.2023.104696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 04/14/2023] Open
Abstract
KDEL receptor (KDELR) is a key protein that recycles escaped ER resident proteins from the Golgi apparatus back to the ER and maintains a dynamic balance between these two organelles in the early secretory pathway. Studies have shown that this retrograde transport pathway is partly regulated by two KDELR-interacting proteins, Acyl-CoA-binding domain-containing 3 (ACBD3), and cyclic AMP-dependent protein kinase A (PKA). However, whether Golgi-localized ACBD3, which was first discovered as a PKA-anchoring protein in mitochondria, directly interacts with PKA at the Golgi and coordinates its signaling in Golgi-to-ER traffic has remained unclear. In this study, we showed that the GOLD domain of ACBD3 directly interacts with the regulatory subunit II (RII) of PKA and effectively recruits PKA holoenzyme to the Golgi. Forward trafficking of proteins from the ER triggers activation of PKA by releasing the catalytic subunit from RII. Furthermore, we determined that depletion of ACBD3 reduces the Golgi fraction of RII, resulting in moderate, but constitutive activation of PKA and KDELR retrograde transport, independent of cargo influx from the ER. Taken together, these data demonstrate that ACBD3 coordinates the protein secretory pathway at the Golgi by facilitating KDELR/PKA-containing protein complex formation.
Collapse
Affiliation(s)
- Jie Jia
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Shuocheng Tang
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Xihua Yue
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Shuaiyang Jing
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Lianhui Zhu
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Chuanting Tan
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jingkai Gao
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yulei Du
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Intaek Lee
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.
| | - Yi Qian
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.
| |
Collapse
|
4
|
Chia S, Leung T, Tan I. Cyclical phosphorylation of LRAP35a and CLASP2 by GSK3β and CK1δ regulates EB1-dependent MT dynamics in cell migration. Cell Rep 2021; 36:109687. [PMID: 34525355 DOI: 10.1016/j.celrep.2021.109687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 07/02/2021] [Accepted: 08/19/2021] [Indexed: 11/30/2022] Open
Abstract
Mammalian cell cytoskeletal reorganization for efficient directional movement requires tight coordination of actomyosin and microtubule networks. In this study, we show that LRAP35a potentiates microtubule stabilization by promoting CLASP2/EB1 interaction besides its complex formation with MRCK/MYO18A for retrograde actin flow. The alternate regulation of these two networks by LRAP35a is tightly regulated by a series of phosphorylation events that dictated its specificity. Sequential phosphorylation of LRAP35a by Protein Kinase A (PKA) and Glycogen Synthase Kinase-3β (GSK3β) initiates the association of LRAP35a with CLASP2, while subsequent binding and further phosphorylation by Casein Kinase 1δ (CK1δ) induce their dissociation, which facilitates LRAP35a/MRCK association in driving lamellar actomyosin flow. Importantly, microtubule dynamics is directly moderated by CK1δ activity on CLASP2 to regulate GSK3β phosphorylation of the SxIP motifs that blocks EB1 binding, an event countered by LRAP35a interaction and its competition for CK1δ activity. Overall this study reveals an essential role for LRAP35a in coordinating lamellar contractility and microtubule polarization in cell migration.
Collapse
Affiliation(s)
- Shumei Chia
- Institute of Molecular and Cell Biology, A(∗)STAR, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; Genome Institute of Singapore, A(∗)STAR, 60 Biopolis Street, #02-01 Genome, Singapore 138672, Singapore; Department of Anatomy, Yong Loo Lin School of Medicine, MD10, 4 Medical Drive, Singapore 117594, Singapore.
| | - Thomas Leung
- Institute of Molecular and Cell Biology, A(∗)STAR, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; Department of Anatomy, Yong Loo Lin School of Medicine, MD10, 4 Medical Drive, Singapore 117594, Singapore
| | - Ivan Tan
- Institute of Molecular and Cell Biology, A(∗)STAR, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; Bioprocessing Technology Institute, A(∗)STAR, 20 Biopolis Way, #06-01, Centros, Singapore 138668, Singapore.
| |
Collapse
|
5
|
Zorzan M, Del Vecchio C, Vogiatzis S, Saccon E, Parolin C, Palù G, Calistri A, Mucignat-Caretta C. Targeting the Regulatory Subunit R2Alpha of Protein Kinase A in Human Glioblastoma through shRNA-Expressing Lentiviral Vectors. Viruses 2021; 13:v13071361. [PMID: 34372567 PMCID: PMC8310305 DOI: 10.3390/v13071361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/25/2021] [Accepted: 07/10/2021] [Indexed: 01/17/2023] Open
Abstract
Glioblastoma is the most malignant and most common form of brain tumor, still today associated with a poor 14-months median survival from diagnosis. Protein kinase A, particularly its regulatory subunit R2Alpha, presents a typical intracellular distribution in glioblastoma cells compared to the healthy brain parenchyma and this peculiarity might be exploited in a therapeutic setting. In the present study, a third-generation lentiviral system for delivery of shRNA targeting the regulatory subunit R2Alpha of protein kinase A was developed. Generated lentiviral vectors are able to induce an efficient and stable downregulation of R2Alpha in different cellular models, including non-stem and stem-like glioblastoma cells. In addition, our data suggest a potential correlation between silencing of the regulatory subunit of protein kinase A and reduced viability of tumor cells, apparently due to a reduction in replication rate. Thus, our findings support the role of protein kinase A as a promising target for novel anti-glioma therapies.
Collapse
|
6
|
US3 Kinase-Mediated Phosphorylation of Tegument Protein VP8 Plays a Critical Role in the Cellular Localization of VP8 and Its Effect on the Lipid Metabolism of Bovine Herpesvirus 1-Infected Cells. J Virol 2019; 93:JVI.02151-18. [PMID: 30626671 DOI: 10.1128/jvi.02151-18] [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: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 02/04/2023] Open
Abstract
Bovine herpesvirus 1 (BoHV-1) infects bovine species, causing respiratory infections, genital disorders and abortions. VP8 is the most abundant tegument protein of BoHV-1 and is critical for virus replication in cattle. In this study, the cellular transport of VP8 in BoHV-1-infected cells and its ability to alter the cellular lipid metabolism were investigated. A viral kinase, US3, was found to be involved in regulating these processes. In the early stages of infection VP8 was localized in the nucleus. Subsequently, presumably after completion of its role in the nucleus, VP8 was translocated to the cytoplasm. When US3 was deleted or the essential US3 phosphorylation site of VP8 was mutated in BoHV-1, the majority of VP8 was localized in the nuclei of infected cells. This suggests that phosphorylation by US3 may be critical for cytoplasmic localization of VP8. Eventually, the cytoplasmic VP8 was accumulated in the cis-Golgi apparatus but not in the trans-Golgi network, implying that VP8 was not involved in virion transport toward and budding from the cell membrane. VP8 caused lipid droplet (LD) formation in the nuclei of transfected cells and increased cellular cholesterol levels. Lipid droplets were not found in the nuclei of BoHV-1-infected cells when VP8 was cytoplasmic in the presence of US3. However, when US3 was deleted or phosphorylation residues in VP8 were mutated, nuclear VP8 and LDs appeared in BoHV-1-infected cells. The total cholesterol level was increased in BoHV-1-infected cells but not in ΔUL47-BoHV-1-infected cells, further supporting a role for VP8 in altering the cellular lipid metabolism during infection.IMPORTANCE Nuclear localization signals (NLSs) and nuclear export signals (NESs) are important elements directing VP8 to the desired locations in the BoHV-1-infected cell. In this study, a critical regulator that switches the nuclear and cytoplasmic localization of VP8 in BoHV-1-infected cells was identified. BoHV-1 used viral kinase US3 to regulate the cellular localization of VP8. Early during BoHV-1 infection VP8 was localized in the nucleus, where it performs various functions; once US3 was expressed, phosphorylated VP8 was cytoplasmic and ultimately accumulated in the cis-Golgi apparatus, presumably to be incorporated into virions. The Golgi localization of VP8 was only observed in virus-infected cells and not in US3-cotransfected cells, suggesting that this is mediated by other viral factors. Interestingly, VP8 was shown to cause increased cholesterol levels, which is a novel function for VP8 and a potential strategy to supply lipid for viral replication.
Collapse
|
7
|
The Golgi architecture and cell sensing. Biochem Soc Trans 2018; 46:1063-1072. [DOI: 10.1042/bst20180323] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/06/2018] [Accepted: 08/21/2018] [Indexed: 12/23/2022]
Abstract
An array of signalling molecules are located at the Golgi apparatus, including phosphoinositides, small GTPases, kinases, and phosphatases, which are linked to multiple signalling pathways. Initially considered to be associated predominantly with membrane trafficking, signalling pathways at the Golgi are now recognised to regulate a diverse range of higher-order functions. Many of these signalling pathways are influenced by the architecture of the Golgi. In vertebrate cells, the Golgi consists of individual stacks fused together into a compact ribbon structure and the function of this ribbon structure has been enigmatic. Notably, recent advances have identified a role for the Golgi ribbon in regulation of cellular processes. Fragmentation of the Golgi ribbon results in modulation of many signalling pathways. Various diseases and disorders, including cancer and neurodegeneration, are associated with the loss of the Golgi ribbon and the appearance of a dispersed fragmented Golgi. Here, we review the emerging theme of the Golgi as a cell sensor and highlight the relationship between the morphological status of the Golgi in vertebrate cells and the modulation of signalling networks.
Collapse
|
8
|
Serebrenik YV, Hellerschmied D, Toure M, López-Giráldez F, Brookner D, Crews CM. Targeted protein unfolding uncovers a Golgi-specific transcriptional stress response. Mol Biol Cell 2018; 29:1284-1298. [PMID: 29851555 PMCID: PMC5994893 DOI: 10.1091/mbc.e17-11-0693] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/26/2018] [Accepted: 03/30/2018] [Indexed: 12/12/2022] Open
Abstract
In eukaryotic cells, organelle-specific stress-response mechanisms are vital for maintaining cellular homeostasis. The Golgi apparatus, an essential organelle of the secretory system, is the major site of protein modification and sorting within a cell and functions as a platform for spatially regulated signaling. Golgi homeostasis mechanisms that regulate organelle structure and ensure precise processing and localization of protein substrates remain poorly understood. Using a chemical biology strategy to induce protein unfolding, we uncover a Golgi-specific transcriptional response. An RNA-sequencing profile of this stress response compared with the current state-of-the-art Golgi stressors, nigericin and xyloside, demonstrates the enhanced precision of Golgi targeting achieved with our system. The data set further reveals previously uncharacterized genes that we find to be essential for Golgi structural integrity. These findings highlight the Golgi's ability to sense misfolded proteins and establish new aspects of Golgi autoregulation.
Collapse
Affiliation(s)
- Yevgeniy V. Serebrenik
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511
| | - Doris Hellerschmied
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511
| | - Momar Toure
- Department of Chemistry, Yale University, New Haven, CT 06511
| | | | - Dennis Brookner
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511
| | - Craig M. Crews
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511
- Department of Chemistry, Yale University, New Haven, CT 06511
- Department of Pharmacology, Yale University, New Haven, CT 06511
| |
Collapse
|
9
|
Mucignat-Caretta C, Denaro L, D'Avella D, Caretta A. Protein Kinase A Distribution Differentiates Human Glioblastoma from Brain Tissue. Cancers (Basel) 2017; 10:cancers10010002. [PMID: 29267253 PMCID: PMC5789352 DOI: 10.3390/cancers10010002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/13/2017] [Accepted: 12/13/2017] [Indexed: 12/17/2022] Open
Abstract
Brain tumor glioblastoma has no clear molecular signature and there is no effective therapy. In rodents, the intracellular distribution of the cyclic AMP (cAMP)-dependent protein kinase (Protein kinase A, PKA) R2Alpha subunit was previously shown to differentiate tumor cells from healthy brain cells. Now, we aim to validate this observation in human tumors. The distribution of regulatory (R1 and R2) and catalytic subunits of PKA was examined via immunohistochemistry and Western blot in primary cell cultures and biopsies from 11 glioblastoma patients. Data were compared with information obtained from 17 other different tumor samples. The R1 subunit was clearly detectable only in some samples. The catalytic subunit was variably distributed in the different tumors. Similar to rodent tumors, all human glioblastoma specimens showed perinuclear R2 distribution in the Golgi area, while it was undetectable outside the tumor. To test the effect of targeting PKA as a therapeutic strategy, the intracellular cyclic AMP concentration was modulated with different agents in four human glioblastoma cell lines. A significant increase in cell death was detected after increasing cAMP levels or modulating PKA activity. These data raise the possibility of targeting the PKA intracellular pathway for the development of diagnostic and/or therapeutic tools for human glioblastoma.
Collapse
Affiliation(s)
- Carla Mucignat-Caretta
- Department of Molecular Medicine, University of Padova, Padova 35131, Italy.
- Biostructures and Biosystems National Institute, Rome 00136, Italy.
| | - Luca Denaro
- Department of Neuroscience, University of Padova, Padova 35131, Italy.
| | - Domenico D'Avella
- Department of Neuroscience, University of Padova, Padova 35131, Italy.
| | - Antonio Caretta
- Biostructures and Biosystems National Institute, Rome 00136, Italy.
- Department of Food and Drug, University of Parma, Parma 43121, Italy.
| |
Collapse
|
10
|
Gosavi P, Gleeson PA. The Function of the Golgi Ribbon Structure - An Enduring Mystery Unfolds! Bioessays 2017; 39. [PMID: 28984991 DOI: 10.1002/bies.201700063] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 08/31/2017] [Indexed: 12/13/2022]
Abstract
The Golgi apparatus in vertebrate cells consists of individual Golgi stacks fused together in a continuous ribbon structure. The ribbon structure per se is not required to mediate the classical functions of this organelle and the relevance of the "ribbon" structure has been a mystery since first identified ultrastructurally in the 1950s. Recent advances recognize a role for the Golgi apparatus in a range of cellular processes, some mediated by signaling networks which are regulated at the Golgi. Here we review the cellular processes and signaling events regulated by the Golgi apparatus and, in particular, explore an emerging theme that the ribbon structure of the Golgi contributes directly to the regulation of these higher order functions.
Collapse
Affiliation(s)
- Prajakta Gosavi
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| | - Paul A Gleeson
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| |
Collapse
|
11
|
Different Golgi ultrastructure across species and tissues: Implications under functional and pathological conditions, and an attempt at classification. Tissue Cell 2017; 49:186-201. [DOI: 10.1016/j.tice.2016.12.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 12/05/2016] [Accepted: 12/05/2016] [Indexed: 02/08/2023]
|
12
|
Tenorio MJ, Luchsinger C, Mardones GA. Protein kinase A activity is necessary for fission and fusion of Golgi to endoplasmic reticulum retrograde tubules. PLoS One 2015; 10:e0135260. [PMID: 26258546 PMCID: PMC4530959 DOI: 10.1371/journal.pone.0135260] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 07/20/2015] [Indexed: 11/18/2022] Open
Abstract
It is becoming increasingly accepted that together with vesicles, tubules play a major role in the transfer of cargo between different cellular compartments. In contrast to our understanding of the molecular mechanisms of vesicular transport, little is known about tubular transport. How signal transduction molecules regulate these two modes of membrane transport processes is also poorly understood. In this study we investigated whether protein kinase A (PKA) activity regulates the retrograde, tubular transport of Golgi matrix proteins from the Golgi to the endoplasmic reticulum (ER). We found that Golgi-to-ER retrograde transport of the Golgi matrix proteins giantin, GM130, GRASP55, GRASP65, and p115 was impaired in the presence of PKA inhibitors. In addition, we unexpectedly found accumulation of tubules containing both Golgi matrix proteins and resident Golgi transmembrane proteins. These tubules were still attached to the Golgi and were highly dynamic. Our data suggest that both fission and fusion of retrograde tubules are mechanisms regulated by PKA activity.
Collapse
Affiliation(s)
- María J. Tenorio
- Instituto de Fisiología, Facultad de Medicina, and Centro Interdisciplinario de Estudios del Sistema Nerviso, Universidad Austral de Chile, Valdivia, Chile
| | - Charlotte Luchsinger
- Instituto de Fisiología, Facultad de Medicina, and Centro Interdisciplinario de Estudios del Sistema Nerviso, Universidad Austral de Chile, Valdivia, Chile
| | - Gonzalo A. Mardones
- Instituto de Fisiología, Facultad de Medicina, and Centro Interdisciplinario de Estudios del Sistema Nerviso, Universidad Austral de Chile, Valdivia, Chile
- * E-mail:
| |
Collapse
|
13
|
Cancino J, Capalbo A, Di Campli A, Giannotta M, Rizzo R, Jung JE, Di Martino R, Persico M, Heinklein P, Sallese M, Luini A. Control systems of membrane transport at the interface between the endoplasmic reticulum and the Golgi. Dev Cell 2014; 30:280-94. [PMID: 25117681 DOI: 10.1016/j.devcel.2014.06.018] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 04/08/2014] [Accepted: 06/23/2014] [Indexed: 10/24/2022]
Abstract
A fundamental property of cellular processes is to maintain homeostasis despite varying internal and external conditions. Within the membrane transport apparatus, variations in membrane fluxes from the endoplasmic reticulum (ER) to the Golgi complex are balanced by opposite fluxes from the Golgi to the ER to maintain homeostasis between the two organelles. Here we describe a molecular device that balances transport fluxes by integrating transduction cascades with the transport machinery. Specifically, ER-to-Golgi transport activates the KDEL receptor at the Golgi, which triggers a cascade that involves Gs and adenylyl cyclase and phosphodiesterase isoforms and then PKA activation and results in the phosphorylation of transport machinery proteins. This induces retrograde traffic to the ER and balances transport fluxes between the ER and Golgi. Moreover, the KDEL receptor activates CREB1 and other transcription factors that upregulate transport-related genes. Thus, a Golgi-based control system maintains transport homeostasis through both signaling and transcriptional networks.
Collapse
Affiliation(s)
- Jorge Cancino
- Istituto di Biochimica delle Proteine, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy; Telethon Institute of Genetics and Medicine, Via Pietro Castellino 111, 80131 Napoli, Italy; Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Quillota 980, Viña del Mar 2520000, Chile.
| | - Anita Capalbo
- Istituto di Biochimica delle Proteine, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy; Telethon Institute of Genetics and Medicine, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Antonella Di Campli
- Istituto di Biochimica delle Proteine, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Monica Giannotta
- Consorzio Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro (Chieti), Italy
| | - Riccardo Rizzo
- Istituto di Biochimica delle Proteine, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy; Telethon Institute of Genetics and Medicine, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Juan E Jung
- Istituto di Biochimica delle Proteine, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy; Istituto di Ricovero e Cura a Carattere Scientifico, Istituto di Ricerca Diagnostica e Nucleare (SDN), 80143 Napoli, Italy
| | - Rosaria Di Martino
- Istituto di Biochimica delle Proteine, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Maria Persico
- Istituto di Biochimica delle Proteine, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy; Istituto di Ricovero e Cura a Carattere Scientifico, Istituto di Ricerca Diagnostica e Nucleare (SDN), 80143 Napoli, Italy
| | - Petra Heinklein
- Institut für Biochemie Charité, Universitätsmedizin Berlin, CrossOver Charitéplatz 1/Sitz, Virchowweg 6, 10117 Berlin, Germany
| | - Michele Sallese
- Consorzio Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro (Chieti), Italy
| | - Alberto Luini
- Istituto di Biochimica delle Proteine, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy; Telethon Institute of Genetics and Medicine, Via Pietro Castellino 111, 80131 Napoli, Italy.
| |
Collapse
|
14
|
Wang Z, Zhang C, Qi RZ. A newly identified myomegalin isoform functions in Golgi microtubule organization and ER-Golgi transport. J Cell Sci 2014; 127:4904-17. [PMID: 25217626 DOI: 10.1242/jcs.155408] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The Golgi of mammalian cells is known to be a major microtubule-organizing site that requires microtubules for its organization and protein trafficking. However, the mechanisms underlying the microtubule organization of the Golgi remain obscure. We used immunoprecipitation coupled with mass spectrometry to identify a widely expressed isoform of the poorly characterized muscle protein myomegalin. This newly identified isoform, myomegalin variant 8 (MMG8), localized predominantly to cis-Golgi networks by interacting with AKAP450 (also known as AKAP9), and this interaction with AKAP450 was required for the stability of both proteins. Disrupting MMG8 expression affected endoplasmic reticulum (ER)-to-Golgi trafficking and caused Golgi fragmentation. Furthermore, MMG8 associated with γ-tubulin complexes and with the microtubule plus-end tracking protein EB1 (also known as MAPRE1), and was required for the Golgi localization of these two molecules. On the Golgi, γ-tubulin complexes mediated microtubule nucleation, whereas EB1 functioned in ER-to-Golgi trafficking. These results indicate that MMG8 participates in Golgi microtubule organization and thereby plays a crucial role in the organization and function of the Golgi.
Collapse
Affiliation(s)
- Zhe Wang
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Chao Zhang
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Robert Z Qi
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| |
Collapse
|
15
|
Jiang Q, Wang L, Guan Y, Xu H, Niu Y, Han L, Wei YP, Lin L, Chu J, Wang Q, Yang Y, Pei L, Wang JZ, Tian Q. Golgin-84-associated Golgi fragmentation triggers tau hyperphosphorylation by activation of cyclin-dependent kinase-5 and extracellular signal-regulated kinase. Neurobiol Aging 2013; 35:1352-63. [PMID: 24368089 DOI: 10.1016/j.neurobiolaging.2013.11.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 09/23/2013] [Accepted: 11/24/2013] [Indexed: 12/16/2022]
Abstract
Tau hyperphosphorylation is a critical event in Alzheimer's disease, in which the neuronal Golgi fragmentation occurs earlier than tau hyperphosphorylation. However, the intrinsic link between Golgi impairment and tau pathology is missing. By electron microscopy and western blotting, we observed in the present study that the neuronal Golgi fragmentation was increased age-dependently with a correlated tau hyperphosphorylation in the brains of C57BL/6 mice aged from 4 to 16 months. Simultaneously, golgin-84 and Golgi reassembly stacking protein 65, 2 important Golgi matrix proteins, were decreased in the brains of elder mice. Further studies in HEK293/tau cells showed that Golgi-disturbing agents, brefeldin A and nocodazole induced tau hyperphosphorylation. Knockdown of golgin-84, not Golgi reassembly stacking protein 65, by small interfering RNA was sufficient to induce tau hyperphosphorylation, while over-expressing golgin-84 arrested the brefeldin A-induced Golgi fragmentation and tau hyperphosphorylation. Finally, we demonstrated that cyclin-dependent kinase-5 and extracellular signal-regulated kinase were activated after golgin-84 knockdown, and simultaneous inhibition of these kinases abolished the golgin-84 deficit-induced tau hyperphosphorylation. These data suggest Golgi fragmentation could be an upstream event triggering tau hyperphosphorylation through golgin-84 deficit-induced activation of cyclin-dependent kinase-5 and extracellular signal-regulated kinase.
Collapse
Affiliation(s)
- Qian Jiang
- Department of Pathology and Pathophysiology, Ultrastructural Pathology Center, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Present address: Department of Dermatology, The First Hospital of Wuhan, Wuhan, China
| | - Lu Wang
- Department of Pathology and Pathophysiology, Ultrastructural Pathology Center, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Guan
- Department of Pathology and Pathophysiology, Ultrastructural Pathology Center, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Xu
- Department of Pathology and Pathophysiology, Ultrastructural Pathology Center, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Niu
- Department of Pathology and Pathophysiology, Ultrastructural Pathology Center, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Han
- Department of Pathology and Pathophysiology, Ultrastructural Pathology Center, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Ping Wei
- Department of Pathology and Pathophysiology, Ultrastructural Pathology Center, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Lin
- Department of Pathology and Pathophysiology, Ultrastructural Pathology Center, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiang Chu
- Department of Pathology and Pathophysiology, Ultrastructural Pathology Center, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qun Wang
- Department of Pathology and Pathophysiology, Ultrastructural Pathology Center, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Yang
- Department of Pathology and Pathophysiology, Ultrastructural Pathology Center, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Pei
- Department of Pathology and Pathophysiology, Ultrastructural Pathology Center, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Department of Pathology and Pathophysiology, Ultrastructural Pathology Center, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qing Tian
- Department of Pathology and Pathophysiology, Ultrastructural Pathology Center, Key Laboratory of Neurological Disease of National Education Ministry and Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
16
|
Regulation of Golgi signaling and trafficking by the KDEL receptor. Histochem Cell Biol 2013; 140:395-405. [DOI: 10.1007/s00418-013-1130-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2013] [Indexed: 12/31/2022]
|
17
|
Lepagnol-Bestel AM, Kvajo M, Karayiorgou M, Simonneau M, Gogos JA. A Disc1 mutation differentially affects neurites and spines in hippocampal and cortical neurons. Mol Cell Neurosci 2013; 54:84-92. [PMID: 23396153 DOI: 10.1016/j.mcn.2013.01.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 01/21/2013] [Accepted: 01/24/2013] [Indexed: 02/05/2023] Open
Abstract
A balanced chromosomal translocation segregating with schizophrenia and affective disorders in a large Scottish family disrupting DISC1 implicated this gene as a susceptibility gene for major mental illness. Here we study neurons derived from a genetically engineered mouse strain with a truncating lesion disrupting the endogenous Disc1 ortholog. We provide a detailed account of the consequences of this mutation on axonal and dendritic morphogenesis as well as dendritic spine development in cultured hippocampal and cortical neurons. We show that the mutation has distinct effects on these two types of neurons, supporting a cell-type specific role of Disc1 in establishing structural connections among neurons. Moreover, using a validated antibody we provide evidence indicating that Disc1 localizes primarily to Golgi apparatus-related vesicles. Our results support the notion that in vitro cultures derived from Disc1(Tm1Kara) mice provide a valuable model for future mechanistic analysis of the cellular and biochemical effects of this mutation, and can thus serve as a platform for drug discovery efforts.
Collapse
Affiliation(s)
- A M Lepagnol-Bestel
- Department of Physiology and Cellular Biophysics, Columbia University, 630 West, 168th Street, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
18
|
Cancino J, Luini A. Signaling Circuits on the Golgi Complex. Traffic 2012; 14:121-34. [DOI: 10.1111/tra.12022] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 10/12/2012] [Accepted: 10/12/2012] [Indexed: 01/21/2023]
|
19
|
Spaeth CS, Spaeth EB, Wilcott RW, Fan JD, Robison T, Bittner GD. Pathways for plasmalemmal repair mediated by PKA, Epac, and cytosolic oxidation in rat B104 cellsin vitroand rat sciatic axonsex vivo. Dev Neurobiol 2012; 72:1399-414. [DOI: 10.1002/dneu.20998] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 11/02/2011] [Indexed: 12/13/2022]
|
20
|
Corda D, Barretta ML, Cervigni RI, Colanzi A. Golgi complex fragmentation in G2/M transition: An organelle-based cell-cycle checkpoint. IUBMB Life 2012; 64:661-70. [PMID: 22730233 DOI: 10.1002/iub.1054] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 04/29/2012] [Indexed: 11/06/2022]
Abstract
In mammalian cells, the Golgi complex is organized into a continuous membranous system known as the Golgi ribbon, which is formed by individual Golgi stacks that are laterally connected by tubular bridges. During mitosis, the Golgi ribbon undergoes extensive fragmentation through a multistage process that is required for its correct partitioning into the daughter cells. Importantly, inhibition of this Golgi disassembly results in cell-cycle arrest at the G2 stage, suggesting that accurate inheritance of the Golgi complex is monitored by a "Golgi mitotic checkpoint." Here, we discuss the mechanisms and regulation of the Golgi ribbon breakdown and briefly comment on how Golgi partitioning may inhibit G2/M transition.
Collapse
Affiliation(s)
- Daniela Corda
- Institute of Protein Biochemistry, National Research Council (CNR), Via Pietro Castellino 111, Naples, Italy.
| | | | | | | |
Collapse
|
21
|
Stenbeck G, Lawrence KM, Albert AP. Hormone-stimulated modulation of endocytic trafficking in osteoclasts. Front Endocrinol (Lausanne) 2012; 3:103. [PMID: 22936925 PMCID: PMC3424527 DOI: 10.3389/fendo.2012.00103] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 08/05/2012] [Indexed: 11/30/2022] Open
Abstract
A precise control of vesicular trafficking is crucial not only for osteoclastic bone resorption, but also for the crosstalk between osteoclasts and osteoblasts, which regulates bone homeostasis. In addition to the release of growth factors and modulators, such as glutamate, flux through the intracellular trafficking routes could also provide the osteoclast with a monitoring function of its resorption activity. To establish the signaling pathways regulating trafficking events in resorbing osteoclasts, we used the bone conserving hormone calcitonin, which has the unique property of inducing osteoclast quiescence. Calcitonin acts through the calcitonin receptor and activates multiple signaling pathways. By monitoring trafficking of a fluorescent low molecular weight probe in mature, bone resorbing osteoclasts we show for the first time that calcitonin blocks endocytosis from the ruffled border by phospholipase C (PLC) activation. Furthermore, we identify a requirement for polyunsaturated fatty acids in endocytic trafficking in osteoclasts. Inhibition of PLC prior to calcitonin treatment restores endocytosis to 75% of untreated rates. This effect is independent of protein kinase C activation and can be mimicked by an increase in intracellular calcium. We thus define an essential role for intracellular calcium levels in the maintenance of endocytosis in osteoclasts.
Collapse
Affiliation(s)
- Gudrun Stenbeck
- Centre for Cell and Chromosome Biology, School of Health Science and Social Care, Brunel UniversityUxbridge, UK
- *Correspondence: Gudrun Stenbeck, Centre for Cell and Chromosome Biology, School of Health Science and Social Care, Heinz Wolff Building, Brunel University, Uxbridge UB8 3PH, UK. e-mail:
| | - Kevin M. Lawrence
- Pharmacology and Cell Physiology, Biomedical Sciences Research Centre, Division of Biomedical Sciences, St George’s, University of LondonLondon, UK
| | - Anthony P. Albert
- Pharmacology and Cell Physiology, Biomedical Sciences Research Centre, Division of Biomedical Sciences, St George’s, University of LondonLondon, UK
| |
Collapse
|
22
|
Abstract
The protein processing and trafficking function of the Golgi is intimately linked to multiple intracellular signaling pathways. Assembly of Golgi trafficking structures and lipid sorting at the Golgi complex is controlled and coordinated by specific phosphoinositide kinases and phosphatases. The intra-Golgi transport machinery is also regulated by kinases belonging to several functionally distinct families, for example, MAP kinase signaling is required for mitotic disassembly of the Golgi. However, the Golgi plays an additional, prominent role in compartmentalizing other signaling cascades that originate at the plasma membrane or at other organelles. This article summarizes recent advances in our understanding of the signaling network that converges at the Golgi.
Collapse
Affiliation(s)
- Peter Mayinger
- Division of Nephrology and Hypertension and Department of Cell and Developmental Biology, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
23
|
Abstract
For growth, survival, communication and homeostasis, cells transport a large number of proteins to the plasma membrane and the extracellular medium by using the secretory pathway. Consequently, to adapt to the surrounding environment and the different intracellular contexts, the secretory pathway needs to accommodate and respond to a plethora of endogenous and exogenous stimuli. It is now well established that several kinases, known to be activated by environmental stimuli, signal from the plasma membrane to the secretory pathway in order to remodel its architecture and modulate the cellular secretion capacity. By contrast, membranes of the early secretory pathway, similar to the endosomal system, can also initiate and modulate signalling cascades, thereby spatially organising cellular signalling and eliciting a different cellular outcome than when signalling is localised to the plasma membrane. This Commentary highlights recent contributions to our understanding of the mutual regulation of the secretory pathway and cellular signalling.
Collapse
Affiliation(s)
- Hesso Farhan
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, Basel, Switzerland.
| | | |
Collapse
|
24
|
Abstract
The Golgi apparatus lies at the heart of the secretory pathway where it receives, modifies and sorts protein cargo to the proper intracellular or extracellular location. Although this secretory function is highly conserved throughout the eukaryotic kingdom, the structure of the Golgi complex is arranged very differently among species. In particular, Golgi membranes in vertebrate cells are integrated into a single compact entity termed the Golgi ribbon that is normally localized in the perinuclear area and in close vicinity to the centrosomes. This organization poses a challenge for cell division when the single Golgi ribbon needs to be partitioned into the two daughter cells. To ensure faithful inheritance in the progeny, the Golgi ribbon is divided in three consecutive steps in mitosis, namely disassembly, partitioning and reassembly. However, the structure of the Golgi ribbon is only present in higher animals and Golgi disassembly during mitosis is not ubiquitous in all organisms. Therefore, there must be unique reasons to build up the Golgi in this particular conformation and to preserve it over generations. In this review, we first highlight the diversity of the Golgi architecture in different organisms and revisit the concept of the Golgi ribbon. Following on, we discuss why the ribbon is needed and how it forms in vertebrate cells. Lastly, we conclude with likely purposes of mitotic ribbon disassembly and further propose mechanisms by which it regulates mitosis.
Collapse
Affiliation(s)
- Jen-Hsuan Wei
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | |
Collapse
|
25
|
Farhan H, Wendeler MW, Mitrovic S, Fava E, Silberberg Y, Sharan R, Zerial M, Hauri HP. MAPK signaling to the early secretory pathway revealed by kinase/phosphatase functional screening. ACTA ACUST UNITED AC 2010; 189:997-1011. [PMID: 20548102 PMCID: PMC2886346 DOI: 10.1083/jcb.200912082] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
To what extent the secretory pathway is regulated by cellular signaling is unknown. In this study, we used RNA interference to explore the function of human kinases and phosphatases in controlling the organization of and trafficking within the secretory pathway. We identified 122 kinases/phosphatases that affect endoplasmic reticulum (ER) export, ER exit sites (ERESs), and/or the Golgi apparatus. Numerous kinases/phosphatases regulate the number of ERESs and ER to Golgi protein trafficking. Among the pathways identified, the Raf-MEK (MAPK/ERK [extracellular signal-regulated kinase] kinase)-ERK cascade, including its regulatory proteins CNK1 (connector enhancer of the kinase suppressor of Ras-1) and neurofibromin, controls the number of ERESs via ERK2, which targets Sec16, a key regulator of ERESs and COPII (coat protein II) vesicle biogenesis. Our analysis reveals an unanticipated complexity of kinase/phosphatase-mediated regulation of the secretory pathway, uncovering a link between growth factor signaling and ER export.
Collapse
Affiliation(s)
- Hesso Farhan
- Biozentrum, Universität Basel, 4056 Basel, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Mavillard F, Hidalgo J, Megias D, Levitsky KL, Velasco A. PKA-mediated Golgi remodeling during cAMP signal transmission. Traffic 2010; 11:90-109. [PMID: 20002352 DOI: 10.1111/j.1600-0854.2009.01007.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cyclic AMP (cAMP)-dependent protein kinase A (PKA) is part of the set of signaling proteins that are stably associated to the cytosolic surface of Golgi membranes in mammalian cells. In principle, Golgi-associated PKA could participate in either signal transduction events and/or the coordination of Golgi transport activities. Here, we show data indicating that although Golgi-associated PKA is activated fast and efficiently during cell stimulation by an extracellular ligand it does not contribute significantly to cAMP signal transmission to the nucleus. Instead, most of the PKA catalytic subunits Calphaderived from the Golgi complex remain localized in the perinuclear cytoplasm where they induce changes in Golgi structural organization. Thus, in stimulated cells the Golgi complex appears collapsed, showing increased colocalization of previously segregated markers and exhibiting merging of different proximal cisternae within a single stack. In contrast, the trans-Golgi network remains as a separate compartment. Consequently, the rate of protein transport is increased whereas glycan processing is not severely affected. This remodeling process requires the presence of PKA activity associated to the Golgi membranes. Together these data indicate that Golgi-associated PKA activity is involved in the adaptation of Golgi dynamic organization to extracellular signaling events.
Collapse
Affiliation(s)
- Fabiola Mavillard
- Department of Cell Biology, Faculty of Biology, University of Seville, Seville, Spain
| | | | | | | | | |
Collapse
|
27
|
Abstract
The mammalian Golgi apparatus is characterized by a ribbon-like organization adjacent to the centrosome during interphase and extensive fragmentation and dispersal away from the centrosome during mitosis. It is not clear whether this dynamic association between the Golgi and centrosome is of functional significance. We discuss recent findings indicating that the Golgi–centrosome relationship may be important for directional protein transport and centrosome positioning, which are both required for cell polarization. We also summarize our current knowledge of the link between Golgi organization and cell cycle progression.
Collapse
Affiliation(s)
- Christine Sütterlin
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA.
| | | |
Collapse
|
28
|
Boal F, Stephens DJ. Specific functions of BIG1 and BIG2 in endomembrane organization. PLoS One 2010; 5:e9898. [PMID: 20360857 PMCID: PMC2845624 DOI: 10.1371/journal.pone.0009898] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 03/04/2010] [Indexed: 11/29/2022] Open
Abstract
Background Transport of molecules from one subcellular compartment to another involves the recruitment of cytosolic coat protein complexes to a donor membrane to concentrate cargo, deform the membrane and ultimately to form an independent carrier. Small-GTP-binding proteins of the Arf family are central to many membrane trafficking events. Arfs are activated by guanine nucleotide exchange factors (GEFs) which results in their recruitment to membranes and subsequent engagement with Arf-effectors, many of which are coat proteins. Among the human BFA-sensitive large Arf-GEFs, the function of the two closely related BIG1 and BIG2 is still not clear, and recent studies have raised the question of functional redundancy between the two proteins. Methodology/Principal Findings Here we have used small-interfering RNA on human cells and a combination of fixed and live-cell imaging to investigate the differential functions of BIG1 and BIG2 in endomembrane organization and function. Importantly, in this direct comparative study, we show discrete functions for BIG1 and BIG2. Our results show that depletion of BIG2 but not of BIG1 induces a tubulation of the recycling endosomal compartment, consistent with a specific role for BIG2 here. In contrast, suppression of BIG1 induces the formation of Golgi mini-stacks still polarized and functional in terms of cargo export. Conclusions A key finding from our work is that suppression of BIG1 expression results in a fragmentation of the Golgi apparatus. Our data indicate that the human BFA-sensitive large Arf-GEFs have non-redundant functions in cell organization and membrane trafficking. BIG1 is required to maintain the normal morphology of the Golgi; BIG2 is important for endosomal compartment integrity and cannot replace the function of BIG1 in Golgi organization.
Collapse
Affiliation(s)
- Frédéric Boal
- Department of Biochemistry, Cell Biology Laboratories, University of Bristol School of Medical Sciences, Bristol, United Kingdom.
| | | |
Collapse
|
29
|
Wojtal KA, Hoekstra D, van Ijzendoorn SCD. cAMP-dependent protein kinase A and the dynamics of epithelial cell surface domains: moving membranes to keep in shape. Bioessays 2008; 30:146-55. [PMID: 18200529 DOI: 10.1002/bies.20705] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cyclic adenosine monophosphate (cAMP) and cAMP-dependent protein kinase A (PKA) are evolutionary conserved molecules with a well-established position in the complex network of signal transduction pathways. cAMP/PKA-mediated signaling pathways are implicated in many biological processes that cooperate in organ development including the motility, survival, proliferation and differentiation of epithelial cells. Cell surface polarity, here defined as the anisotropic organisation of cellular membranes, is a critical parameter for most of these processes. Changes in the activity of cAMP/PKA elicit a variety of effects on intracellular membrane dynamics, including membrane sorting and trafficking. One of the most intriguing aspects of cAMP/PKA signaling is its evolutionary conserved abundance on the one hand and its precise spatial-temporal actions on the other. Here, we review recent developments with regard to the role of cAMP/PKA in the regulation of intracellular membrane trafficking in relation to the dynamics of epithelial surface domains.
Collapse
Affiliation(s)
- Kacper A Wojtal
- Division of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
30
|
Immunocytochemical analysis of cyclic AMP receptor proteins in the developing rat parotid gland. Arch Oral Biol 2008; 53:429-36. [PMID: 18187105 DOI: 10.1016/j.archoralbio.2007.11.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2007] [Revised: 10/23/2007] [Accepted: 11/26/2007] [Indexed: 11/20/2022]
Abstract
UNLABELLED Previous studies showed that regulatory subunits of type II cyclic AMP-dependent protein kinase (RII) are present in adult rat parotid acinar cells, and are secreted into saliva. If the synthesis and intracellular distribution of RII exhibit developmental specificity, then RII can be an indicator of secretory and regulatory activity of salivary glands. OBJECTIVE To determine the expression and distribution of RII in the rat parotid at specific ages representing defined developmental stages. METHODS Parotid glands of fetal, neonatal and adult rats were prepared for morphologic and immunocytochemical study. The cellular distribution of RII was studied using light microscopic immunogold silver staining with anti-RII, and its intracellular distribution using electron microscopic immunogold labeling. RESULTS In utero, parotid RII levels were low; 5-18 days after birth, labeling of secretory granules and cytoplasm rose to a peak, followed by a rapid decrease in both compartments at 25 days. At 60 days, granule labeling increased to levels near those at 18 days, whereas cytoplasmic labeling remained low. Nuclear labeling was highest during the first 3 weeks after birth, and then declined. CONCLUSIONS The higher nuclear and cytoplasmic labeling during the neonatal period may reflect RII involvement in acinar cell differentiation. The accumulation of RII in secretory granules is similar to the pattern of the major salivary proteins, amylase and PSP. The redistribution of RII in these compartments during development may reflect changing gene expression patterns, and may be useful for identification of genetic or metabolic abnormalities.
Collapse
|
31
|
Langhans M, Hawes C, Hillmer S, Hummel E, Robinson DG. Golgi regeneration after brefeldin A treatment in BY-2 cells entails stack enlargement and cisternal growth followed by division. PLANT PHYSIOLOGY 2007; 145:527-38. [PMID: 17704232 PMCID: PMC2048719 DOI: 10.1104/pp.107.104919] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2007] [Accepted: 08/02/2007] [Indexed: 05/05/2023]
Abstract
Brefeldin A (BFA) treatment stops secretion and leads to the resorption of much of the Golgi apparatus into the endoplasmic reticulum. This effect is reversible upon washing out the drug, providing a situation for studying Golgi biogenesis. In this investigation Golgi regeneration in synchronized tobacco BY-2 cells was followed by electron microscopy and by the immunofluorescence detection of ARF1, which localizes to the rims of Golgi cisternae and serves as an indicator of COPI vesiculation. Beginning as clusters of vesicles that are COPI positive, mini-Golgi stacks first become recognizable 60 min after BFA washout. They continue to increase in terms of numbers and length of cisternae for a further 90 min before overshooting the size of control Golgi stacks. As a result, increasing numbers of dividing Golgi stacks were observed 120 min after BFA washout. BFA-regeneration experiments performed on cells treated with BFA (10 microg mL(-1)) for only short periods (30-45 min) showed that the formation of ER-Golgi hybrid structures, once initiated by BFA treatment, is an irreversible process, the further incorporation of Golgi membranes into the ER continuing during a subsequent drug washout. Application of the protein kinase A inhibitor H-89, which effectively blocks the reassembly of the Golgi apparatus in mammalian cells, also prevented stack regeneration in BY-2 cells, but only at very high, almost toxic concentrations (>200 microm). Our data suggest that under normal conditions mitosis-related Golgi stack duplication may likely occur via cisternal growth followed by fission.
Collapse
Affiliation(s)
- Markus Langhans
- Department of Cell Biology, Heidelberg Institute for Plant Sciences, University of Heidelberg, 69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
32
|
Sun AQ, Ponamgi VM, Boyer JL, Suchy FJ. Membrane trafficking of the human organic anion-transporting polypeptide C (hOATPC). Pharm Res 2007; 25:463-74. [PMID: 17641954 DOI: 10.1007/s11095-007-9399-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2007] [Accepted: 05/03/2007] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The human organic anion transporting polypeptide C (OATPC) is one of the major transport proteins involved in the enterohepatic circulation of bile salts and plays an important role in vectorial transport of organic anions and drugs across hepatocytes. MATERIALS AND METHODS In this study, the effects of biological reagents on the membrane localization of OATPC were investigated by confocal microscopy and estrone-3-sulfate transport. RESULTS Our results demonstrated that the functional membrane expression of fluorescent chimera OATPC-GFP was achieved in non-polarized (COS7 and HEK293) and polarized (MDCK) cells. Both brefeldin A (a Golgi complex disruptor) and bafilomycin A1 (an inhibitor of vacuolar H+-ATPase) treatment significantly decreased the polarized membrane trafficking and markedly reduced the uptake of estrone-3-sulfate ( approximately 40-90%) in OATPC-GFP transfected cells, suggesting that membrane sorting of hOATPC-GFP was mediated by Golgi complex and vacuolar H+-ATPase-related vesicle transport pathways. Treatment with 8-Br-cAMP (a cAMP analog) stimulated OATPC-GFP membrane localization and enhanced estrone-3-sulfate uptake by approximately 20%. The protein kinase A (PKA) inhibitors (H89 and KT5720), but not a PKG inhibitor, blocked the polarized membrane expression of OATPC-GFP and reduced estrone-3-sulfate transport activity. The simultaneous treatment of cells with PKA activator/inhibitor and bafilomycin A1 demonstrated that bafilomycin A1 did not change the effects of 8-Br-cAMP and H89 on the membrane localization of OATPC-GFP compared with the use of 8-Br-cAMP and H89 alone. DISCUSSION These data suggest that a cAMP-PKA sensitive membrane sorting pathway for OATPC-GFP is independent of the vacuolar H+-ATPase associated (bafilomycin A1 sensitive) vesicle mediated membrane sorting pathway. In contrast, with combined treatment with brefeldin A, neither the PKA-activator (8-Br-cAMP) nor the inhibitor (H89) further altered the plasma membrane expression and transport activity of OATPC-GFP compared with brefeldin A treatment alone. These data suggest that the cAMP-PKA regulation of OATPC membrane expression involves the Golgi complex. When the Golgi apparatus was disrupted by brefeldin A treatment, the effects of cAMP-PKA on the Golgi-to-basolateral surface sorting process of OATPC was also diminished. In summary, the plasma membrane localization of human OATPC is mediated by Golgi complex and vacuolar H+-ATPase vesicle mediated membrane sorting pathways. cAMP-PKA regulates sorting process through the Golgi complex but not the vacuolar H+-ATPase associated vesicular pathway.
Collapse
Affiliation(s)
- An-Qiang Sun
- Department of Pediatrics, Mount Sinai School of Medicine, PO Box 1664, One Gustave L. Levy Place, New York, NY 10029-6574, USA.
| | | | | | | |
Collapse
|
33
|
Kirk SJ, Ward TH. COPII under the microscope. Semin Cell Dev Biol 2007; 18:435-47. [PMID: 17693103 DOI: 10.1016/j.semcdb.2007.07.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2007] [Revised: 07/05/2007] [Accepted: 07/09/2007] [Indexed: 11/19/2022]
Abstract
Transport through the secretory pathway begins with COPII regulation of ER export. Driven by the Sar1 GTPase cycle, cytosolic COPII proteins exchange on and off the membrane at specific sites on the ER to regulate cargo exit. Here recent developments in COPII research are discussed, particularly the use of live-cell imaging, which has revealed surprising insights into the coat's role. The seemingly static ER exit sites are in fact highly dynamic, and the ability to visualise trafficking processes in intact living cells has highlighted the adaptable nature of COPII in cargo transport and the emerging roles of auxiliary factors.
Collapse
Affiliation(s)
- Semra J Kirk
- Immunology Unit, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK.
| | | |
Collapse
|
34
|
Kim HS, Takahashi M, Matsuo K, Ono Y. Recruitment of CG-NAP to the Golgi apparatus through interaction with dynein-dynactin complex. Genes Cells 2007; 12:421-34. [PMID: 17352745 DOI: 10.1111/j.1365-2443.2007.01055.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The structural organization and position of the Golgi apparatus are highly regulated by microtubule cytoskeleton and microtubule motor proteins. The mechanisms linking these proteins to the Golgi apparatus remain elusive. Here, we found that centrosome and Golgi-localized PKN associated protein (CG-NAP) was localized to the Golgi apparatus in a microtubule-dependent manner. Microtubule-binding experiments revealed that CG-NAP possessed two microtubule-binding domains. We also found that CG-NAP was well co-localized with cytoplasmic dynein subunits during recovery from the on-ice treatment of cells that induced dissociation of CG-NAP from the Golgi. Similar co-localization was observed during recovery from the acetate treatment, which has been reported to inhibit the dynein-mediated transport. CG-NAP was co-immunoprecipitated with a dynactin subunit p150(Glued). Expressing the p150(Glued)-binding region of CG-NAP fused with mitochondria-targeting sequence induced recruitment of mitochondria to the pericentriolar area, suggesting that this region interacts with functional cytoplasmic dynein in vivo. Moreover, over-expression of this region caused fragmentation of the Golgi similar to that of dynamitin. These results suggest that CG-NAP is recruited to the minus ends of microtubules by interacting with cytoplasmic dynein, thereby localizes to the Golgi apparatus in a microtubule-dependent manner and possibly involved in the formation of the Golgi near the centrosomes.
Collapse
Affiliation(s)
- Hon-Song Kim
- Graduate School of Science and Technology, Kobe University, Kobe 657-8501, Japan
| | | | | | | |
Collapse
|
35
|
Morgado-Díaz JA, Montesano G, De Souza Fernandes S, Redondo PA, Fernandes de Souza W, Albuquerque-Xavier AC, Leve F, Tanaka MN, Martins de Araujo W, Oliveira SS, Benchimol M, De Souza W. Golgi complex disassembly caused by light-activated calphostin C involves MAPK and PKA. Tissue Cell 2007; 39:161-9. [PMID: 17412380 DOI: 10.1016/j.tice.2007.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We examined the participation of MAPK and PKA in the Golgi complex disassembly caused by light-activated Calphostin C in HT-29 cells. When these cells were incubated with Calphostin C, fragmentation and dispersal of the Golgi complex was observed as assessed by immunofluorescence microscopy. Electron microscopy analysis showed that clusters of vesicles and large tubule-vesicular membrane structures, resembling the Golgi remnants present in mitotic cells, substituted the Golgi stacks. In addition, Calphostin C treatment caused inhibition of the endocytic route. We confirmed that the Golgi disassembly was not due to PKC inhibition, and suggested, based on the use of specific inhibitors, that other kinases are involved. It was shown that pretreatment with PD98059 and H-89, both inhibitors of MAPK and PKA, respectively, prior to incubation with Calphostin C, caused blockade of the Golgi disassembly, as well as the inhibition of the endocytic pathway caused by this drug. This finding supports the existence of a novel mechanism by which MAPK and PKA may regulate the Golgi breakdown caused by Calphostin C in HT-29 cells.
Collapse
Affiliation(s)
- J A Morgado-Díaz
- Grupo de Biologia Estrutural, Divisão de Biologia Celular, Centro de Pesquisas, Instituto Nacional de Câncer, Rio de Janeiro, RJ 20231-050, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kuroda F, Moss J, Vaughan M. Regulation of brefeldin A-inhibited guanine nucleotide-exchange protein 1 (BIG1) and BIG2 activity via PKA and protein phosphatase 1gamma. Proc Natl Acad Sci U S A 2007; 104:3201-6. [PMID: 17360629 PMCID: PMC1805514 DOI: 10.1073/pnas.0611696104] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Brefeldin A-inhibited guanine nucleotide-exchange proteins (GEPs) BIG1 and BIG2 activate ADP-ribosylation factor (ARF) GTPases, which are required for vesicular trafficking. Both molecules contain one or more sites for binding protein kinase A, i.e., A kinase-anchoring protein (AKAP) sequences. Elevation of cell cAMP caused PKA-catalyzed phosphorylation and nuclear accumulation of BIG1 but not BIG2. We then asked whether BIG1 phosphorylation altered its GEP activity. Incubation of BIG1 or BIG2 with PKA catalytic subunits and ATP resulted in retardation of their electrophoretic migration, consistent with PKA phosphorylation. Okadaic acid inhibits many protein phosphatases, including protein phosphatase 1 (PP1) and PP2A, that can reverse PKA-catalyzed phosphorylation. Incubation of HepG2 cells with okadaic acid caused concentration-dependent accumulation of presumably phosphorylated BIG1 and BIG2 with decreased mobility, which was increased by subsequent incubation in vitro with specific recombinant phosphatases, PP1gamma > PP2A >> PP1alpha. For assays of GEP activity, BIG1 and BIG2 were immunoprecipitated from cells that had been depleted, respectively, of BIG2 and BIG1 by using specific siRNA. GEP activity of each was significantly decreased after incubation with recombinant PKA plus ATP and restored by incubation with PP1gamma. In agreement with a role for PP1gamma in regulation of BIG, endogenous PP1gamma, but not PP1alpha or beta, was immunoprecipitated with BIG1 or BIG2 from microsomal fractions. All observations are consistent with the effects of BIG1 and BIG2 phosphorylation on vesicular trafficking, via alterations in ARF activation and regulatory roles for cAMP, PKA, and PP1gamma in ARF activation by BIG1 and BIG2.
Collapse
Affiliation(s)
- Fuminobu Kuroda
- Pulmonary–Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1434
| | - Joel Moss
- Pulmonary–Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1434
| | - Martha Vaughan
- Pulmonary–Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1434
- *To whom correspondence should be addressed at:
Pulmonary–Critical Care Medicine Branch, Building 10/5N307, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1434. E-mail:
| |
Collapse
|