1
|
Su M, Radhakrishnan A, Yan Y, Tian Y, Zheng H, M’Saad O, Graham M, Coleman J, Goder JND, Liu X, Zhang Y, Bewersdorf J, Rothman JE. The Golgi Rim is a Precise Tetraplex of Golgin Proteins that Can Self-Assemble into Filamentous Bands. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.645134. [PMID: 40196516 PMCID: PMC11974933 DOI: 10.1101/2025.03.27.645134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Golgin proteins have long been suspected to be organizers of the Golgi stack. Using three-dimensional super-resolution microscopy, we comprehensively localize the human golgin family at the rim of the Golgi apparatus at 10-20 nm resolution in situ. Unexpectedly, we find that the golgins are precisely organized into a tetraplex with four discrete layers, each containing a specific set of rim golgins. We observe no golgins inside the stack between its membrane-bound cisternae. Biochemically characterizing most of the golgins as isolated proteins, we find that they form anti-parallel dimers and further self-assemble into bands of multi-micron-long filaments. Based on our findings, we propose an "outside-in" physical model, the Golgin Organizer Hypothesis, in which the Golgi stack of cisternae and its overall ribbon morphology directly result from bending circumferential bands of rim golgin filaments onto a membrane surface, explaining stack formation without the need for special "stacking proteins."
Collapse
Affiliation(s)
- Maohan Su
- Department of Cell Biology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Abhijith Radhakrishnan
- Department of Cell Biology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | | | - Yuan Tian
- Department of Cell Biology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Hong Zheng
- Department of Cell Biology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Ons M’Saad
- Department of Cell Biology, School of Medicine, Yale University, New Haven, CT 06520, USA
- Current address: Panluminate Inc., New Haven, CT 06520, USA
| | - Morven Graham
- Department of Cell Biology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Jeff Coleman
- Department of Cell Biology, School of Medicine, Yale University, New Haven, CT 06520, USA
- Nanobiology Institute, Yale University, West Haven, CT 06516, USA
| | - Jean N. D. Goder
- Department of Cell Biology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Xinran Liu
- Department of Cell Biology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Yongdeng Zhang
- Department of Cell Biology, School of Medicine, Yale University, New Haven, CT 06520, USA
- Current address: School of Life Sciences, Westlake University, Hangzhou 310024, China
| | - Joerg Bewersdorf
- Department of Cell Biology, School of Medicine, Yale University, New Haven, CT 06520, USA
- Nanobiology Institute, Yale University, West Haven, CT 06516, USA
| | - James E. Rothman
- Department of Cell Biology, School of Medicine, Yale University, New Haven, CT 06520, USA
- Nanobiology Institute, Yale University, West Haven, CT 06516, USA
| |
Collapse
|
2
|
Wang J, Niu S, Hu X, Li T, Liu S, Tu Y, Shang Z, Zhao L, Xu P, Lin J, Chen L, Billadeau DD, Jia D. Trans-Golgi network tethering factors regulate TBK1 trafficking and promote the STING-IFN-I pathway. Cell Discov 2025; 11:23. [PMID: 40097395 PMCID: PMC11914254 DOI: 10.1038/s41421-024-00763-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/22/2024] [Indexed: 03/19/2025] Open
Abstract
The cGAS-STING pathway mediates the innate immune response to cytosolic DNA, contributing to surveillance against microbial invasion or cellular damage. Once activated, STING recruits TBK1 at the trans-Golgi network (TGN), which in turn phosphorylates IRF3 to induce type I interferon (IFN-I) expression. In contrast to STING, little is known about how TBK1 is transported to the TGN for activation. Here, we show that multiple TGN tethering factors, a group of proteins involved in vesicle capturing, are indispensable for STING-IFN-I signaling. Deletion of TBC1D23, a recently reported tethering factor, in mice impairs the STING-IFN-I signaling, but with insignificant effect on STING-NF-κB signaling. Mechanistically, TBC1D23 interacts with TBK1 via the WASH complex subunit FAM21 and promotes its endosome-to-TGN translocation. Furthermore, multiple TGN tethering factors were reduced in aged mice and senescent fibroblasts. In summary, our study uncovers that TGN tethering factors are key regulators of the STING-IFN-I signaling and suggests that their reduction in senescence may produce aberrant STING signaling.
Collapse
Affiliation(s)
- Jinrui Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Shenghui Niu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Xiao Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Tianxing Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Shengduo Liu
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yingfeng Tu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Zehua Shang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Lin Zhao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Pinglong Xu
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingwen Lin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Lu Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Daniel D Billadeau
- Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Da Jia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China.
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Ferreira AM, Silva-Álvarez V, Kraev I, Uysal-Onganer P, Lange S. Extracellular vesicles and citrullination signatures are novel biomarkers in sturgeon (Acipenser gueldenstaedtii) during chronic stress due to seasonal temperature challenge. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109974. [PMID: 39426640 DOI: 10.1016/j.fsi.2024.109974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/03/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
Acipenser gueldenstaedtii is one of the most cultured sturgeon species worldwide and of considerable economic value for caviar production. There are though considerable challenges around chronic stress responses due to increased summer temperatures, impacting sturgeons' immune responses and their susceptibility to opportunistic infections. The identification of molecular and cellular pathways involved in stress responses may contribute to identifying novel biomarkers reflective of fish health status, crucial for successful sturgeon aquaculture. Protein citrullination is a calcium-catalysed post-translational modification caused by peptidylarginine deiminases (PADs), altering target protein function and affecting protein interactions in physiological and pathobiological processes. PADs can also modulate extracellular vesicle (EVs) profiles, which play critical roles in cellular communication, via transport of their cargoes (proteins, including post-translationally modified proteins, genetic material and micro-RNAs). This study identified differences in EV signatures, and citrullinated proteins in sera from winter and summer farmed sturegeons. EVs were significantly elevated in sera of the summer chronically stressed group. The citrullinated proteins and associated gene ontology (GO) pathways in sera and serum-EVs of chronically heat stressed A. gueldenstaedtii, showed some changes, with specific citrullinated serum protein targets including alpa-2-macroglobulin, alpha globin, calcium-dependent secretion activator, ceruloplasmin, chemokine XC receptor, complement C3 isoforms, complement C9, plectin, selenoprotein and vitellogenin. In serum-EVs, citrullinated protein cargoes identified only in the chronically stressed summer group included alpha-1-antiproteinase, apolipoprotein B-100, microtubule actin crosslinking factor and histone H3. Biological gene ontology (GO) pathways related to citrullinated serum proteins in the chronically stressed group were associated with innate and adaptive immune responses, stress responses and metabolic processes. In serum-EVs of the heat-stressed group the citrullinome associated with various metabolic GO pathways. In addition to modified citrullinated protein content, Serum-EVs from the stressed summer group showed significantly increased levels of the inflammatory associated miR-155 and the hypoxia-associated miR-210, but significantly reduced levels of the growth-associated miR-206. Our findings highlight roles for protein citrullination and EV signatures in response to chronic heat stress in A. gueldenstaedtii, indicating a trade-off in immunity versus growth and may be of value for sturgeon aquaculture.
Collapse
Affiliation(s)
- Ana María Ferreira
- Laboratorio de Inmunología, Instituto de Química Biológica, Facultad de Ciencias, Instituto de Higiene, UdelaR, Uruguay; Área de Inmunología, Departamento de Biociencias, Facultad de Química, Instituto de Higiene, UdelaR, Uruguay.
| | - Valeria Silva-Álvarez
- Laboratorio de Inmunología, Instituto de Química Biológica, Facultad de Ciencias, Instituto de Higiene, UdelaR, Uruguay; Área de Inmunología, Departamento de Biociencias, Facultad de Química, Instituto de Higiene, UdelaR, Uruguay.
| | - Igor Kraev
- Electron Microscopy Suite, Faculty of Science, Technology, Engineering and Mathematics, Open University, Milton Keynes MK7 6AA, UK.
| | - Pinar Uysal-Onganer
- Cancer Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK.
| | - Sigrun Lange
- Pathobiology and Extracellular Vesicles Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK.
| |
Collapse
|
4
|
Yao Y, Xu Y, Yu L, Xue T, Xiao Z, Tin P, Fung H, Ma H, Yun J, Yam JWP. NHE7 upregulation potentiates the uptake of small extracellular vesicles by enhancing maturation of macropinosomes in hepatocellular carcinoma. Cancer Commun (Lond) 2024; 44:251-272. [PMID: 38152992 PMCID: PMC10876205 DOI: 10.1002/cac2.12515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 12/29/2023] Open
Abstract
BACKGROUND Small extracellular vesicles (sEVs) mediate intercellular communication that contributes to hepatocellular carcinoma (HCC) progression via multifaceted pathways. The success of cell entry determines the effect of sEV on recipient cells. Here, we aimed to delineate the mechanisms underlying the uptake of sEV in HCC. METHODS Macropinocytosis was examined by the ability of cells to internalize dextran and sEV. Macropinocytosis was analyzed in Na(+)/H(+) exchanger 7 (NHE7)-knockdown and -overexpressing cells. The properties of cells were studied using functional assays. pH biosensor was used to evaluate the intracellular and endosomal pH. The expression of NHE7 in patients' liver tissues was examined by immunofluorescent staining. Inducible silencing of NHE7 in established tumors was performed to reveal the therapeutic potential of targeting NHE7. RESULTS The data revealed that macropinocytosis controlled the internalization of sEVs and their oncogenic effect on recipient cells. It was found that metastatic HCC cells exhibited the highest efficiency of sEV uptake relative to normal liver cells and non-metastatic HCC cells. Attenuation of macropinocytic activity by 5-(N-ethyl-N-isopropyl)-amiloride (EIPA) limited the entry of sEVs and compromised cell aggressiveness. Mechanistically, we delineated that high level of NHE7, a sodium-hydrogen exchanger, alkalized intracellular pH and acidized endosomal pH, leading to the maturation of macropinosomes. Inducible inhibition of NHE7 in established tumors developed in mice delayed tumor development and suppressed lung metastasis. Clinically, NHE7 expression was upregulated and linked to dismal prognosis of HCC. CONCLUSIONS This study advances the understanding that NHE7 enhances sEV uptake by macropinocytosis to promote the malignant properties of HCC cells. Inhibition of sEV uptake via macropinocytosis can be exploited as a treatment alone or in combination with conventional therapeutic approaches for HCC.
Collapse
Affiliation(s)
- Yue Yao
- Department of PathologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongP. R. China
- Department of Endocrinology and MetabolismSecond Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjingP. R. China
| | - Yi Xu
- Department of PathologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongP. R. China
- Department of Hepatopancreatobiliary SurgerySecond Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjingP. R. China
- State Key Laboratory of Oncology in South ChinaCancer Center of Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
| | - Liang Yu
- Department of PathologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongP. R. China
- Department of Hepatopancreatobiliary SurgerySecond Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjingP. R. China
| | - Ting‐Mao Xue
- Department of PathologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongP. R. China
- Department of Hepatobiliary Surgery IIZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Zhi‐Jie Xiao
- Scientific Research CenterThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongP. R. China
| | - Pui‐Chi Tin
- Department of PathologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongP. R. China
| | - Hiu‐Ling Fung
- Department of PathologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongP. R. China
| | - Hoi‐Tang Ma
- Department of PathologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongP. R. China
- State Key Laboratory of Liver ResearchThe University of Hong KongHong KongP. R. China
| | - Jing‐Ping Yun
- Department of PathologyCancer Center of Sun Yat‐sen UniversityGuangzhouGuangdongP. R. China
| | - Judy Wai Ping Yam
- Department of PathologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongP. R. China
- State Key Laboratory of Liver ResearchThe University of Hong KongHong KongP. R. China
| |
Collapse
|
5
|
Pillsbury CE, Dougan J, Rabe JL, Fonseca JA, Zhou C, Evans AN, Abukharma H, Ichoku O, Gonzalez-Flamenco G, Park SI, Aljudi A, DeRyckere D, Castellino SM, Rafiq S, Langermann S, Liu LN, Henry CJ, Porter CC. Siglec-15 Promotes Evasion of Adaptive Immunity in B-cell Acute Lymphoblastic Leukemia. CANCER RESEARCH COMMUNICATIONS 2023; 3:1248-1259. [PMID: 37465593 PMCID: PMC10351425 DOI: 10.1158/2767-9764.crc-23-0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/28/2023] [Accepted: 06/20/2023] [Indexed: 07/20/2023]
Abstract
Siglec-15 (Sig15) has been implicated as an immune checkpoint expressed in solid tumor-infiltrating macrophages and is being targeted in clinical trials with mAbs to normalize the tumor immune microenvironment and stimulate antitumor immunity. However, the role of Sig15 in hematologic malignancies remains undefined. Sig15 mRNA and protein expression levels in hematologic malignancies were determined from publicly available databases, cell lines, and primary patient samples. Human B-cell acute lymphoblastic leukemia (B-ALL) cell lines were used to identify signaling pathways involved in the regulation of Sig15 expression. Secreted/soluble Sig15 and cytokine levels were measured from the plasma of children with leukemia and healthy controls. Knockdown and knockout of Siglec15 in a murine model of B-ALL was used to evaluate the effect of leukemia-derived Sig15 on the immune response to leukemia. We observed pathologic overexpression of Sig15 in a variety of hematologic malignancies, including primary B-ALL samples. This overexpression was driven by NFκB activation, which also increased the surface localization of Sig15. Secreted/soluble Sig15 was found to circulate at elevated levels in the plasma of children with B-ALL and correlated with an immune-suppressive cytokine milieu. Genetic inhibition of Sig15 in murine B-ALL promoted clearance of the leukemia by the immune system and a marked reversal of the immune-privileged leukemia bone marrow niche, including expanded early effector CD8+ T cells and reduction of immunosuppressive cytokines. Thus, Sig15 is a novel, potent immunosuppressive molecule active in leukemia that may be targeted therapeutically to activate T lymphocytes against leukemia cells. Significance We demonstrate that Sig15 is overexpressed in hematologic malignancies driven by NFκB, is required for immune evasion in a mouse model of leukemia, and, for the first time, that it circulates at high levels in the plasma of children with leukemia.
Collapse
Affiliation(s)
- Claire E. Pillsbury
- Cancer Biology Program, Laney Graduate School, Emory University, Atlanta, Georgia
| | - Jodi Dougan
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Jennifer L. Rabe
- Molecular Biology Program, University of Colorado Denver, Aurora, Colorado
| | - Jairo A. Fonseca
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Chengjing Zhou
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Alyssa N. Evans
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | | | | | | | - Sunita I. Park
- Clinical Laboratory, Children's Healthcare of Atlanta, Atlanta, Georgia
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia
| | - Ahmed Aljudi
- Clinical Laboratory, Children's Healthcare of Atlanta, Atlanta, Georgia
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia
| | - Deborah DeRyckere
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Sharon M. Castellino
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Sarwish Rafiq
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | | | | | - Curtis J. Henry
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Christopher C. Porter
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| |
Collapse
|
6
|
PKD-dependent PARP12-catalyzed mono-ADP-ribosylation of Golgin-97 is required for E-cadherin transport from Golgi to plasma membrane. Proc Natl Acad Sci U S A 2022; 119:2026494119. [PMID: 34969853 PMCID: PMC8740581 DOI: 10.1073/pnas.2026494119] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2021] [Indexed: 11/18/2022] Open
Abstract
Adenosine diphosphate (ADP)-ribosylation is a posttranslational modification involved in key regulatory events catalyzed by ADP-ribosyltransferases (ARTs). Substrate identification and localization of the mono-ADP-ribosyltransferase PARP12 at the trans-Golgi network (TGN) hinted at the involvement of ARTs in intracellular traffic. We find that Golgin-97, a TGN protein required for the formation and transport of a specific class of basolateral cargoes (e.g., E-cadherin and vesicular stomatitis virus G protein [VSVG]), is a PARP12 substrate. PARP12 targets an acidic cluster in the Golgin-97 coiled-coil domain essential for function. Its mutation or PARP12 depletion, delays E-cadherin and VSVG export and leads to a defect in carrier fission, hence in transport, with consequent accumulation of cargoes in a trans-Golgi/Rab11-positive intermediate compartment. In contrast, PARP12 does not control the Golgin-245-dependent traffic of cargoes such as tumor necrosis factor alpha (TNFα). Thus, the transport of different basolateral proteins to the plasma membrane is differentially regulated by Golgin-97 mono-ADP-ribosylation by PARP12. This identifies a selective regulatory mechanism acting on the transport of Golgin-97- vs. Golgin-245-dependent cargoes. Of note, PARP12 enzymatic activity, and consequently Golgin-97 mono-ADP-ribosylation, depends on the activation of protein kinase D (PKD) at the TGN during traffic. PARP12 is directly phosphorylated by PKD, and this is essential to stimulate PARP12 catalytic activity. PARP12 is therefore a component of the PKD-driven regulatory cascade that selectively controls a major branch of the basolateral transport pathway. We propose that through this mechanism, PARP12 contributes to the maintenance of E-cadherin-mediated cell polarity and cell-cell junctions.
Collapse
|
7
|
Brumfield A, Chaudhary N, Molle D, Wen J, Graumann J, McGraw TE. Insulin-promoted mobilization of GLUT4 from a perinuclear storage site requires RAB10. Mol Biol Cell 2021; 32:57-73. [PMID: 33175605 PMCID: PMC8098823 DOI: 10.1091/mbc.e20-06-0356] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/30/2020] [Accepted: 11/04/2020] [Indexed: 12/05/2022] Open
Abstract
Insulin controls glucose uptake into muscle and fat cells by inducing a net redistribution of glucose transporter 4 (GLUT4) from intracellular storage to the plasma membrane (PM). The TBC1D4-RAB10 signaling module is required for insulin-stimulated GLUT4 translocation to the PM, although where it intersects GLUT4 traffic was unknown. Here we demonstrate that TBC1D4-RAB10 functions to control GLUT4 mobilization from a trans-Golgi network (TGN) storage compartment, establishing that insulin, in addition to regulating the PM proximal effects of GLUT4-containing vesicles docking to and fusion with the PM, also directly regulates the behavior of GLUT4 deeper within the cell. We also show that GLUT4 is retained in an element/domain of the TGN from which newly synthesized lysosomal proteins are targeted to the late endosomes and the ATP7A copper transporter is translocated to the PM by elevated copper. Insulin does not mobilize ATP7A nor does copper mobilize GLUT4, and RAB10 is not required for copper-elicited ATP7A mobilization. Consequently, GLUT4 intracellular sequestration and mobilization by insulin is achieved, in part, through utilizing a region of the TGN devoted to specialized cargo transport in general rather than being specific for GLUT4. Our results define the GLUT4-containing region of the TGN as a sorting and storage site from which different cargo are mobilized by distinct signals through unique molecular machinery.
Collapse
Affiliation(s)
| | - Natasha Chaudhary
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Dorothee Molle
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Jennifer Wen
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Johannes Graumann
- Weill Cornell Medical College in Qatar, Education City, 24144 Doha, State of Qatar
| | - Timothy E. McGraw
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
- Department of Cardiothoracic Surgery, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
8
|
The ADP-ribosylation factor-like small GTPase FgArl1 participates in growth, pathogenicity and DON production in Fusarium graminearum. Fungal Biol 2020; 124:969-980. [PMID: 33059848 DOI: 10.1016/j.funbio.2020.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 08/08/2020] [Accepted: 08/20/2020] [Indexed: 01/04/2023]
Abstract
Fusarium graminearum is the main pathogen of Fusarium head blight (FHB) in wheat and related species, which causes serious production decreases and economic losses and produces toxins such as deoxynivalenol (DON), which endangers the health of humans and livestock. Vesicle transport is a basic physiological process required for cell survival in eukaryotes. Many regulators of vesicle transport are reported to be involved in the pathogenicity of fungi. In yeast and mammalian cells, the ADP-ribosylation factor-like small GTPase Arl1 and its orthologs are involved in regulating vesicular trafficking, cytoskeletal reorganization and other significant biological processes. However, the role of Arl1 in F. graminearum is not well understood. In this study, we characterized the Arl1-homologous protein FgArl1 in F. graminearum and showed that FgArl1 is located in the trans-Golgi apparatus. The deletion of FgARL1 resulted in a significant decrease in vegetative growth and pathogenicity. Further analyses of the ΔFgarl1 mutant revealed defects in the production of DON. Taken together, these results indicate that FgArl1 is important in the development and pathogenicity of F. graminearum.
Collapse
|
9
|
Ireland SC, Huang H, Zhang J, Li J, Wang Y. Hydrogen peroxide induces Arl1 degradation and impairs Golgi-mediated trafficking. Mol Biol Cell 2020; 31:1931-1942. [PMID: 32583744 PMCID: PMC7525819 DOI: 10.1091/mbc.e20-01-0063] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/02/2020] [Accepted: 06/09/2020] [Indexed: 12/13/2022] Open
Abstract
Reactive oxygen species (ROS)-induced oxidative stress has been associated with diseases such as amyotrophic lateral sclerosis, stroke, and cancer. While the effect of ROS on mitochondria and endoplasmic reticulum (ER) has been well documented, its consequence on the Golgi apparatus is less well understood. In this study, we characterized the Golgi structure and function in HeLa cells after exposure to hydrogen peroxide (H2O2), a reagent commonly used to introduce ROS to cells. Treatment of cells with 1 mM H2O2 for 10 min resulted in the degradation of Arl1 and dissociation of GRIP domain-containing proteins Golgin-97 and Golgin-245 from the trans-Golgi. This effect could be rescued by treatment of cells with a ROS scavenger N-acetyl cysteine or protease inhibitors. Structurally, H2O2 treatment reduced the number of cisternal membranes per Golgi stack, suggesting a loss of trans-Golgi cisternae. Functionally, H2O2 treatment inhibited both anterograde and retrograde protein transport, consistent with the loss of membrane tethers on the trans-Golgi cisternae. This study revealed membrane tethers at the trans-Golgi as novel specific targets of ROS in cells.
Collapse
Affiliation(s)
- Stephen C. Ireland
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085
| | - Haoran Huang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085
| | - Jianchao Zhang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085
| | - Jie Li
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085
- Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI 48109-1085
| |
Collapse
|
10
|
Ishida M, Bonifacino JS. ARFRP1 functions upstream of ARL1 and ARL5 to coordinate recruitment of distinct tethering factors to the trans-Golgi network. J Cell Biol 2019; 218:3681-3696. [PMID: 31575603 PMCID: PMC6829661 DOI: 10.1083/jcb.201905097] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/09/2019] [Accepted: 08/27/2019] [Indexed: 12/22/2022] Open
Abstract
SNARE-mediated fusion of endosome-derived transport carriers with the trans-Golgi network (TGN) depends on the concerted action of two types of tethering factors: long coiled-coil tethers of the golgin family, and the heterotetrameric complex GARP. Whereas the golgins mediate long-distance capture of the carriers, GARP promotes assembly of the SNAREs. It remains to be determined, however, how the functions of these tethering factors are coordinated. Herein we report that the ARF-like (ARL) GTPase ARFRP1 functions upstream of two other ARL GTPases, ARL1 and ARL5, which in turn recruit golgins and GARP, respectively, to the TGN. We also show that this mechanism is essential for the delivery of retrograde cargos to the TGN. Our findings thus demonstrate that ARFRP1 is a master regulator of retrograde-carrier tethering to the TGN. The coordinated recruitment of distinct tethering factors by a bifurcated GTPase cascade may be paradigmatic of other vesicular fusion events within the cell.
Collapse
Affiliation(s)
- Morié Ishida
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Juan S Bonifacino
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| |
Collapse
|
11
|
Sztul E, Chen PW, Casanova JE, Cherfils J, Dacks JB, Lambright DG, Lee FJS, Randazzo PA, Santy LC, Schürmann A, Wilhelmi I, Yohe ME, Kahn RA. ARF GTPases and their GEFs and GAPs: concepts and challenges. Mol Biol Cell 2019; 30:1249-1271. [PMID: 31084567 PMCID: PMC6724607 DOI: 10.1091/mbc.e18-12-0820] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/26/2019] [Accepted: 03/11/2019] [Indexed: 12/12/2022] Open
Abstract
Detailed structural, biochemical, cell biological, and genetic studies of any gene/protein are required to develop models of its actions in cells. Studying a protein family in the aggregate yields additional information, as one can include analyses of their coevolution, acquisition or loss of functionalities, structural pliability, and the emergence of shared or variations in molecular mechanisms. An even richer understanding of cell biology can be achieved through evaluating functionally linked protein families. In this review, we summarize current knowledge of three protein families: the ARF GTPases, the guanine nucleotide exchange factors (ARF GEFs) that activate them, and the GTPase-activating proteins (ARF GAPs) that have the ability to both propagate and terminate signaling. However, despite decades of scrutiny, our understanding of how these essential proteins function in cells remains fragmentary. We believe that the inherent complexity of ARF signaling and its regulation by GEFs and GAPs will require the concerted effort of many laboratories working together, ideally within a consortium to optimally pool information and resources. The collaborative study of these three functionally connected families (≥70 mammalian genes) will yield transformative insights into regulation of cell signaling.
Collapse
Affiliation(s)
- Elizabeth Sztul
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Pei-Wen Chen
- Department of Biology, Williams College, Williamstown, MA 01267
| | - James E. Casanova
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908
| | - Jacqueline Cherfils
- Laboratoire de Biologie et Pharmacologie Appliquée, CNRS and Ecole Normale Supérieure Paris-Saclay, 94235 Cachan, France
| | - Joel B. Dacks
- Division of Infectious Disease, Department of Medicine, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - David G. Lambright
- Program in Molecular Medicine and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Amherst, MA 01605
| | - Fang-Jen S. Lee
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | | | - Lorraine C. Santy
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802
| | - Annette Schürmann
- German Institute of Human Nutrition, 85764 Potsdam-Rehbrücke, Germany
| | - Ilka Wilhelmi
- German Institute of Human Nutrition, 85764 Potsdam-Rehbrücke, Germany
| | - Marielle E. Yohe
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892
| | - Richard A. Kahn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322-3050
| |
Collapse
|
12
|
Bhatt JM, Hancock W, Meissner JM, Kaczmarczyk A, Lee E, Viktorova E, Ramanadham S, Belov GA, Sztul E. Promiscuity of the catalytic Sec7 domain within the guanine nucleotide exchange factor GBF1 in ARF activation, Golgi homeostasis, and effector recruitment. Mol Biol Cell 2019; 30:1523-1535. [PMID: 30943106 PMCID: PMC6724685 DOI: 10.1091/mbc.e18-11-0711] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The integrity of the Golgi and trans-Golgi network (TGN) is disrupted by brefeldin A (BFA), which inhibits the Golgi-localized BFA-sensitive factor (GBF1) and brefeldin A-inhibited guanine nucleotide-exchange factors (BIG1 and BIG2). Using a cellular replacement assay to assess GBF1 functionality without interference from the BIGs, we show that GBF1 alone maintains Golgi architecture; facilitates secretion; activates ADP-ribosylation factor (ARF)1, 3, 4, and 5; and recruits ARF effectors to Golgi membranes. Unexpectedly, GBF1 also supports TGN integrity and recruits numerous TGN-localized ARF effectors. The impact of the catalytic Sec7 domain (Sec7d) on GBF1 functionality was assessed by swapping it with the Sec7d from ARF nucleotide-binding site opener (ARNO)/cytohesin-2, a plasma membrane GEF reported to activate all ARFs. The resulting chimera (GBF1-ARNO-GBF1 [GARG]) targets like GBF1, supports Golgi/TGN architecture, and facilitates secretion. However, unlike GBF1, GARG activates all ARFs (including ARF6) at the Golgi/TGN and recruits additional ARF effectors to the Golgi/TGN. Our results have general implications: 1) GEF's targeting is independent of Sec7d, but Sec7d influence the GEF substrate specificity and downstream effector events; 2) all ARFs have access to all membranes, but are restricted in their distribution by the localization of their activating GEFs; and 3) effector association with membranes requires the coincidental presence of activated ARFs and specific membrane identifiers.
Collapse
Affiliation(s)
- Jay M Bhatt
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - William Hancock
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Justyna M Meissner
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Aneta Kaczmarczyk
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Eunjoo Lee
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Ekaterina Viktorova
- Department of Veterinary Medicine, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
| | - Sasanka Ramanadham
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294.,Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - George A Belov
- Department of Veterinary Medicine, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742
| | - Elizabeth Sztul
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
13
|
Despres J, Ramdani Y, di Giovanni M, Bénard M, Zahid A, Montero-Hadjadje M, Yvergnaux F, Saguet T, Driouich A, Follet-Gueye ML. Replicative senescence of human dermal fibroblasts affects structural and functional aspects of the Golgi apparatus. Exp Dermatol 2019; 28:922-932. [DOI: 10.1111/exd.13886] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/14/2018] [Accepted: 01/02/2019] [Indexed: 01/02/2023]
Affiliation(s)
- Julie Despres
- Laboratoire Glycobiologie et Matrice Extracellulaire Végétale; Normandie Université; UNIROUEN, Fédération de recherche Normandie-Végétal - FED 4277, GDR CNRS 3711, COSM'ACTIFS; Mont-Saint-Aignan France
- Bioeurope; Groupe SOLABIA; Route d'OulinsAnet France
| | - Yasmina Ramdani
- Laboratoire Glycobiologie et Matrice Extracellulaire Végétale; Normandie Université; UNIROUEN, Fédération de recherche Normandie-Végétal - FED 4277, GDR CNRS 3711, COSM'ACTIFS; Mont-Saint-Aignan France
| | - Marine di Giovanni
- Cell Imaging Platform (PRIMACEN-IRIB); Normandie Université; UNIROUEN; Mont-Saint-Aignan France
| | - Magalie Bénard
- Cell Imaging Platform (PRIMACEN-IRIB); Normandie Université; UNIROUEN; Mont-Saint-Aignan France
| | - Abderrakib Zahid
- Laboratoire Glycobiologie et Matrice Extracellulaire Végétale; Normandie Université; UNIROUEN, Fédération de recherche Normandie-Végétal - FED 4277, GDR CNRS 3711, COSM'ACTIFS; Mont-Saint-Aignan France
| | - Maité Montero-Hadjadje
- Laboratoire de Différenciation et Communication Neuronale et Neuroendocrine; Institut de Recherche et d'Innovation Biomédicale de Normandie; Normandie Univ; UNIROUEN; INSERM U1239; Rouen France
| | | | | | - Azeddine Driouich
- Laboratoire Glycobiologie et Matrice Extracellulaire Végétale; Normandie Université; UNIROUEN, Fédération de recherche Normandie-Végétal - FED 4277, GDR CNRS 3711, COSM'ACTIFS; Mont-Saint-Aignan France
- Cell Imaging Platform (PRIMACEN-IRIB); Normandie Université; UNIROUEN; Mont-Saint-Aignan France
| | - Marie-Laure Follet-Gueye
- Laboratoire Glycobiologie et Matrice Extracellulaire Végétale; Normandie Université; UNIROUEN, Fédération de recherche Normandie-Végétal - FED 4277, GDR CNRS 3711, COSM'ACTIFS; Mont-Saint-Aignan France
- Cell Imaging Platform (PRIMACEN-IRIB); Normandie Université; UNIROUEN; Mont-Saint-Aignan France
| |
Collapse
|
14
|
Cui Y, Yang Z, Teasdale RD. A role of GCC88 in the retrograde transport of CI-M6PR and the maintenance of lysosomal activity. Cell Biol Int 2019; 43:1234-1244. [PMID: 30791178 DOI: 10.1002/cbin.11118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/17/2019] [Indexed: 11/10/2022]
Abstract
GCC88 is a golgin coiled-coil protein at the trans-Golgi (TGN) that functions as a tethering factor for the endosome-derived retrograde transport vesicles. Here, we demonstrate that GCC88 is required for the endosome-to-TGN retrograde transport of the cation-independent mannose 6-phosphate receptor (CI-M6PR). The knockout of GCC88 perturbs the retrieval of CI-M6PR and decreases its cellular level at the steady state, which causes the improper processing of newly synthesized cathepsin-D, a lysosomal hydrolase dependent on CI-M6PR for its delivery to lysosomes. At the whole cell level, the knockout of GCC88 reduces the lysosomal proteolytic capacity but does not impair of the efficiency of autophagy within these cells.
Collapse
Affiliation(s)
- Yi Cui
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Zhe Yang
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Rohan D Teasdale
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
15
|
Abstract
The role of the Golgi apparatus in carcinogenesis still remains unclear. A number of structural and functional cis-, medial-, and trans-Golgi proteins as well as a complexity of metabolic pathways which they mediate may indicate a central role of the Golgi apparatus in the development and progression of cancer. Pleiotropy of cellular function of the Golgi apparatus makes it a "metabolic heart" or a relay station of a cell, which combines multiple signaling pathways involved in carcinogenesis. Therefore, any damage to or structural abnormality of the Golgi apparatus, causing its fragmentation and/or biochemical dysregulation, results in an up- or downregulation of signaling pathways and may in turn promote tumor progression, as well as local nodal and distant metastases. Three alternative or parallel models of spatial and functional Golgi organization within tumor cells were proposed: (1) compacted Golgi structure, (2) normal Golgi structure with its increased activity, and (3) the Golgi fragmentation with ministacks formation. Regardless of the assumed model, the increased activity of oncogenesis initiators and promoters with inhibition of suppressor proteins results in an increased cell motility and migration, increased angiogenesis, significantly activated trafficking kinetics, proliferation, EMT induction, decreased susceptibility to apoptosis-inducing factors, and modulating immune response to tumor cell antigens. Eventually, this will lead to the increased metastatic potential of cancer cells and an increased risk of lymph node and distant metastases. This chapter provided an overview of the current state of knowledge of selected Golgi proteins, their role in cytophysiology as well as potential involvement in tumorigenesis.
Collapse
|
16
|
Tie HC, Ludwig A, Sandin S, Lu L. The spatial separation of processing and transport functions to the interior and periphery of the Golgi stack. eLife 2018; 7:41301. [PMID: 30499774 PMCID: PMC6294550 DOI: 10.7554/elife.41301] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/30/2018] [Indexed: 12/15/2022] Open
Abstract
It is unclear how the two principal functions of the Golgi complex, processing and transport, are spatially organized. Studying such spatial organization by optical imaging is challenging, partially due to the dense packing of stochastically oriented Golgi stacks. Using super-resolution microscopy and markers such as Giantin, we developed a method to identify en face and side views of individual nocodazole-induced Golgi mini-stacks. Our imaging uncovered that Golgi enzymes preferentially localize to the cisternal interior, appearing as a central disk or inner-ring, whereas components of the trafficking machinery reside at the periphery of the stack, including the cisternal rim. Interestingly, conventional secretory cargos appeared at the cisternal interior during their intra-Golgi trafficking and transiently localized to the cisternal rim before exiting the Golgi. In contrast, bulky cargos were found only at the rim. Our study therefore directly demonstrates the spatial separation of processing and transport functions within the Golgi complex.
Collapse
Affiliation(s)
- Hieng Chiong Tie
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Alexander Ludwig
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Sara Sandin
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,NTU Institute of Structural Biology, Nanyang Technological University, Singapore, Singapore
| | - Lei Lu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
17
|
Taneja TK, Ma D, Kim BY, Welling PA. Golgin-97 Targets Ectopically Expressed Inward Rectifying Potassium Channel, Kir2.1, to the trans-Golgi Network in COS-7 Cells. Front Physiol 2018; 9:1070. [PMID: 30123141 PMCID: PMC6085455 DOI: 10.3389/fphys.2018.01070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 07/17/2018] [Indexed: 12/27/2022] Open
Abstract
The inward rectifying potassium channel, Kir2.1, is selected as cargo at the trans-Golgi network (TGN) for export to the cell surface through a unique signal-dependent interaction with the AP1 clathrin-adaptor, but it is unknown how the channel is targeted at earlier stages in the secretory pathway for traffic to the TGN. Here we explore a mechanism. A systematic screen of Golgi tethers identified Golgin-97 as a Kir2.1 binding partner. In vitro protein-interaction studies revealed the interaction is direct, occurring between the GRIP domain of Golgin-97 and the cytoplasmic domain of Kir2.1. Imaging and interaction studies in COS-7 cells suggest that Golgi-97 binds to the channel en route through the Golgi. RNA interference-mediated knockdown of Golgin-97 prevented exit of Kir2.1 from the Golgi. These observations identify Golgin-97 as a Kir2.1 binding partner that is required for targeting the channel to the TGN. Based on our studies in COS-7 cells, we propose Golgi-97 facilitates formation of AP1-dependent export carriers for Kir2.1 by coupling anterograde delivery of Kir2.1 with retrograde recycling of AP-1 containing endosomes to the TGN.
Collapse
Affiliation(s)
- Tarvinder K Taneja
- Department of Physiology, Maryland Center for Kidney Discovery, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Donghui Ma
- Department of Physiology, Maryland Center for Kidney Discovery, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Bo Y Kim
- Department of Physiology, Maryland Center for Kidney Discovery, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Paul A Welling
- Department of Physiology, Maryland Center for Kidney Discovery, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| |
Collapse
|
18
|
Hsu RM, Zhong CY, Wang CL, Liao WC, Yang C, Lin SY, Lin JW, Cheng HY, Li PY, Yu CJ. Golgi tethering factor golgin-97 suppresses breast cancer cell invasiveness by modulating NF-κB activity. Cell Commun Signal 2018; 16:19. [PMID: 29703230 PMCID: PMC5923015 DOI: 10.1186/s12964-018-0230-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 04/13/2018] [Indexed: 12/13/2022] Open
Abstract
Background Golgin-97 is a tethering factor in the trans-Golgi network (TGN) and is crucial for vesicular trafficking and maintaining cell polarity. However, the significance of golgin-97 in human diseases such as cancer remains unclear. Methods We searched for a potential role of golgin-97 in cancers using Kaplan-Meier Plotter (http://kmplot.com) and Oncomine (www.oncomine.org) datasets. Specific functions of golgin-97 in migration and invasion were examined in golgin-97-knockdown and golgin-97-overexpressing cells. cDNA microarray, pathway analysis and qPCR were used to identify gene profiles regulated by golgin-97. The role of golgin-97 in NF-κB signaling pathway was examined by using subcellular fractionation, luciferase reporter assay, western blot analysis and immunofluorescence assay (IFA). Results We found that low expression of golgin-97 correlated with poor overall survival of cancer patients and was associated with invasiveness in breast cancer cells. Golgin-97 knockdown promoted cell migration and invasion, whereas re-expression of golgin-97 restored the above phenotypes in breast cancer cells. Microarray and pathway analyses revealed that golgin-97 knockdown induced the expression of several invasion-promoting genes that were transcriptionally regulated by NF-κB p65. Mechanistically, golgin-97 knockdown significantly reduced IκBα protein levels and activated NF-κB, whereas neither IκBα levels nor NF-κB activity was changed in TGN46- or GCC185-knockdown cells. Conversely, golgin-97 overexpression suppressed NF-κB activity and restored the levels of IκBα in golgin-97-knockdown cells. Interestingly, the results of Golgi-disturbing agent treatment revealed that the loss of Golgi integrity was not involved in the NF-κB activation induced by golgin-97 knockdown. Moreover, both TGN-bound and cytosolic golgin-97 inhibited NF-κB activation, indicating that golgin-97 functions as an NF-κB suppressor regardless of its subcellular localization. Conclusion Our results collectively demonstrate a novel and suppressive role of golgin-97 in cancer invasiveness. We also provide a new avenue for exploring the relationship between the TGN, golgin-97 and NF-κB signaling in tumor progression. Electronic supplementary material The online version of this article (10.1186/s12964-018-0230-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rae-Mann Hsu
- Department of Cell and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Cai-Yan Zhong
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Liang Wang
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Pulmonary Oncology and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Wei-Chao Liao
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan.,Department of Otolaryngology - Head & Neck Surgery, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan.,Center for General Education, Chang Gung University, Taoyuan, Taiwan
| | - Chi Yang
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Shih-Yu Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jia-Wei Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsiao-Yun Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Po-Yu Li
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Jung Yu
- Department of Cell and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Division of Pulmonary Oncology and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan. .,Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
19
|
Gilbert CE, Sztul E, Machamer CE. Commonly used trafficking blocks disrupt ARF1 activation and the localization and function of specific Golgi proteins. Mol Biol Cell 2018; 29:937-947. [PMID: 29467256 PMCID: PMC5896932 DOI: 10.1091/mbc.e17-11-0622] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cold temperature blocks used to synchronize protein trafficking inhibit GBF1 function, leading to a decrease in ARF1-GTP levels and mislocalization of the ARF1 effector golgin-160. Several other, but not all, Golgi proteins including ARL1 also mislocalize. ARF1 activity and golgin-160 localization require more than 30 min to recover from these blocks. ADP-ribosylation factor (ARF) proteins are key regulators of the secretory pathway. ARF1, through interacting with its effectors, regulates protein trafficking by facilitating numerous events at the Golgi. One unique ARF1 effector is golgin-160, which promotes the trafficking of only a specific subset of cargo proteins through the Golgi. While studying this role of golgin-160, we discovered that commonly used cold temperature blocks utilized to synchronize cargo trafficking (20 and 16°C) caused golgin-160 dispersal from Golgi membranes. Here, we show that the loss of golgin-160 localization correlates with a decrease in the levels of activated ARF1, and that golgin-160 dispersal can be prevented by expression of a GTP-locked ARF1 mutant. Overexpression of the ARF1 activator Golgi brefeldin A–resistant guanine nucleotide exchange factor 1 (GBF1) did not prevent golgin-160 dispersal, suggesting that GBF1 may be nonfunctional at lower temperatures. We further discovered that several other Golgi resident proteins had altered localization at lower temperatures, including proteins recruited by ARF-like GTPase 1 (ARL1), a small GTPase that also became dispersed in the cold. Although cold temperature blocks are useful for synchronizing cargo trafficking through the Golgi, our data indicate that caution must be taken when interpreting results from these assays.
Collapse
Affiliation(s)
- Catherine E Gilbert
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Elizabeth Sztul
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35924
| | - Carolyn E Machamer
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
20
|
Gosavi P, Houghton FJ, McMillan PJ, Hanssen E, Gleeson PA. The Golgi ribbon in mammalian cells negatively regulates autophagy by modulating mTOR activity. J Cell Sci 2018; 131:jcs.211987. [PMID: 29361552 DOI: 10.1242/jcs.211987] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 12/20/2017] [Indexed: 12/25/2022] Open
Abstract
In vertebrates, individual Golgi stacks are joined into a compact ribbon structure; however, the relevance of a ribbon structure has been elusive. Here, we exploit the finding that the membrane tether of the trans-Golgi network, GCC88 (encoded by GCC1), regulates the balance between Golgi mini-stacks and the Golgi ribbon. Loss of Golgi ribbons in stable cells overexpressing GCC88 resulted in compromised mechanistic target of rapamycin (mTOR) signaling and a dramatic increase in LC3-II-positive autophagosomes, whereas RNAi-mediated depletion of GCC88 restored the Golgi ribbon and reduced autophagy. mTOR was absent from dispersed Golgi mini-stacks whereas recruitment of mTOR to lysosomes was unaffected. We show that the Golgi ribbon is a site for localization and activation of mTOR, a process dependent on the ribbon structure. We demonstrate a strict temporal sequence of fragmentation of Golgi ribbon, loss of Golgi mTOR and subsequent increased autophagy. Golgi ribbon fragmentation has been reported in various neurodegenerative diseases and we demonstrate the potential relevance of our findings in neuronal cells using a model of neurodegeneration. Overall, this study highlights a role for the Golgi ribbon in pathways central to cellular homeostasis.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Prajakta Gosavi
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Fiona J Houghton
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Paul J McMillan
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia.,Biological Optical Microscopy Platform, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Eric Hanssen
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia.,Advanced Microscopy Facility, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, 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, Melbourne, Victoria 3010, Australia
| |
Collapse
|
21
|
Abstract
ADP-ribosylation factors (Arfs) and ADP-ribosylation factor-like proteins (Arls) are highly conserved small GTPases that function as main regulators of vesicular trafficking and cytoskeletal reorganization. Arl1, the first identified member of the large Arl family, is an important regulator of Golgi complex structure and function in organisms ranging from yeast to mammals. Together with its effectors, Arl1 has been shown to be involved in several cellular processes, including endosomal trans-Golgi network and secretory trafficking, lipid droplet and salivary granule formation, innate immunity and neuronal development, stress tolerance, as well as the response of the unfolded protein. In this Commentary, we provide a comprehensive summary of the Arl1-dependent cellular functions and a detailed characterization of several Arl1 effectors. We propose that involvement of Arl1 in these diverse cellular functions reflects the fact that Arl1 is activated at several late-Golgi sites, corresponding to specific molecular complexes that respond to and integrate multiple signals. We also provide insight into how the GTP-GDP cycle of Arl1 is regulated, and highlight a newly discovered mechanism that controls the sophisticated regulation of Arl1 activity at the Golgi complex.
Collapse
Affiliation(s)
- Chia-Jung Yu
- Department of Cell and Molecular Biology, College of Medicine, Chang Gung University, Linkou, Tao-Yuan 33302, Taiwan.,Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Tao-Yuan 33305, Taiwan
| | - Fang-Jen S Lee
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan .,Department of Medical Research, National Taiwan University Hospital, Taipei 10002, Taiwan
| |
Collapse
|
22
|
Kumar M, Kaur S, Nazir A, Tripathi RK. HIV-1 Nef binds with human GCC185 protein and regulates mannose 6 phosphate receptor recycling. Biochem Biophys Res Commun 2016; 474:137-145. [DOI: 10.1016/j.bbrc.2016.04.086] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 04/18/2016] [Indexed: 01/28/2023]
|
23
|
Cheung PYP, Pfeffer SR. Transport Vesicle Tethering at the Trans Golgi Network: Coiled Coil Proteins in Action. Front Cell Dev Biol 2016; 4:18. [PMID: 27014693 PMCID: PMC4791371 DOI: 10.3389/fcell.2016.00018] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 02/29/2016] [Indexed: 12/14/2022] Open
Abstract
The Golgi complex is decorated with so-called Golgin proteins that share a common feature: a large proportion of their amino acid sequences are predicted to form coiled-coil structures. The possible presence of extensive coiled coils implies that these proteins are highly elongated molecules that can extend a significant distance from the Golgi surface. This property would help them to capture or trap inbound transport vesicles and to tether Golgi mini-stacks together. This review will summarize our current understanding of coiled coil tethers that are needed for the receipt of transport vesicles at the trans Golgi network (TGN). How do long tethering proteins actually catch vesicles? Golgi-associated, coiled coil tethers contain numerous binding sites for small GTPases, SNARE proteins, and vesicle coat proteins. How are these interactions coordinated and are any or all of them important for the tethering process? Progress toward understanding these questions and remaining, unresolved mysteries will be discussed.
Collapse
Affiliation(s)
- Pak-Yan P Cheung
- Department of Biochemistry, Stanford University School of Medicine Stanford, CA, USA
| | - Suzanne R Pfeffer
- Department of Biochemistry, Stanford University School of Medicine Stanford, CA, USA
| |
Collapse
|
24
|
Cheung PYP, Limouse C, Mabuchi H, Pfeffer SR. Protein flexibility is required for vesicle tethering at the Golgi. eLife 2015; 4. [PMID: 26653856 PMCID: PMC4721967 DOI: 10.7554/elife.12790] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 12/13/2015] [Indexed: 01/27/2023] Open
Abstract
The Golgi is decorated with coiled-coil proteins that may extend long distances to help vesicles find their targets. GCC185 is a trans Golgi-associated protein that captures vesicles inbound from late endosomes. Although predicted to be relatively rigid and highly extended, we show that flexibility in a central region is required for GCC185’s ability to function in a vesicle tethering cycle. Proximity ligation experiments show that that GCC185’s N-and C-termini are within <40 nm of each other on the Golgi. In physiological buffers without fixatives, atomic force microscopy reveals that GCC185 is shorter than predicted, and its flexibility is due to a central bubble that represents local unwinding of specific sequences. Moreover, 85% of the N-termini are splayed, and the splayed N-terminus can capture transport vesicles in vitro. These unexpected features support a model in which GCC185 collapses onto the Golgi surface, perhaps by binding to Rab GTPases, to mediate vesicle tethering.
Collapse
Affiliation(s)
| | - Charles Limouse
- Department of Applied Physics, Stanford University, Stanford, United States
| | - Hideo Mabuchi
- Department of Applied Physics, Stanford University, Stanford, United States
| | - Suzanne R Pfeffer
- Department of Biochemistry, Stanford University School of Medicine, Stanford, United States
| |
Collapse
|
25
|
Huang LH, Lee WC, You ST, Cheng CC, Yu CJ. Arfaptin-1 negatively regulates Arl1-mediated retrograde transport. PLoS One 2015; 10:e0118743. [PMID: 25789876 PMCID: PMC4366199 DOI: 10.1371/journal.pone.0118743] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 01/11/2015] [Indexed: 12/24/2022] Open
Abstract
The small GTPase Arf-like protein 1 (Arl1) is well known for its role in intracellular vesicular transport at the trans-Golgi network (TGN). In this study, we used differential affinity chromatography combined with mass spectrometry to identify Arf-interacting protein 1b (arfaptin-1b) as an Arl1-interacting protein and characterized a novel function for arfaptin-1 (including the arfaptin-1a and 1b isoforms) in Arl1-mediated retrograde transport. Using a Shiga-toxin subunit B (STxB) transportation assay, we demonstrated that knockdown of arfaptin-1 accelerated the retrograde transport of STxB from the endosome to the Golgi apparatus, whereas Arl1 knockdown inhibited STxB transport compared with control cells. Arfaptin-1 overexpression, but not an Arl1 binding-defective mutant (arfaptin-1b-F317A), consistently inhibited STxB transport. Exogenous arfaptin-1 expression did not interfere with the localization of the Arl1-interacting proteins golgin-97 and golgin-245 to the TGN and vice versa. Moreover, we found that the N-terminal region of arfaptin-1 was involved in the regulation of retrograde transport. Our results show that arfaptin-1 acts as a negative regulator in Arl1-mediated retrograde transport and suggest that different functional complexes containing Arl1 form in distinct microdomains and are responsible for different functions.
Collapse
Affiliation(s)
- Lien-Hung Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Wei-Chung Lee
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Shu-Ting You
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Chen Cheng
- Department of Cell and Molecular Biology, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Chia-Jung Yu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Department of Cell and Molecular Biology, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Tao-Yuan, Taiwan
- * E-mail:
| |
Collapse
|
26
|
Abstract
The ARF-like (ARL) proteins, within the ARF family, are a collection of functionally diverse GTPases that share extensive (>40 %) identity with the ARFs and each other and are assumed to share basic mechanisms of regulation and a very incompletely documented degree of overlapping regulators. At least four ARLs were already present in the last eukaryotic common ancestor, along with one ARF, and these have been expanded to >20 members in mammals. We know little about the majority of these proteins so our review will focus on those about which the most is known, including ARL1, ARL2, ARL3, ARL4s, ARL6, ARL13s, and ARFRP1. From this fragmentary information we extract some generalizations and conclusions regarding the sources and extent of specificity and functions of the ARLs.
Collapse
Affiliation(s)
- Alfred Wittinghofer
- Max-Planck-Institute of Molecular Physiology, Dortmund, Nordrhein-Westfalen Germany
| |
Collapse
|
27
|
Li CC, Wu TS, Huang CF, Jang LT, Liu YT, You ST, Liou GG, Lee FJS. GTP-binding-defective ARL4D alters mitochondrial morphology and membrane potential. PLoS One 2012; 7:e43552. [PMID: 22927989 PMCID: PMC3424131 DOI: 10.1371/journal.pone.0043552] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 07/23/2012] [Indexed: 12/01/2022] Open
Abstract
ARL4D, ARL4A, and ARL4C are closely related members of the ADP-ribosylation factor/ARF-like protein (ARF/ARL) family of GTPases. All three ARL4 proteins contain nuclear localization signals (NLSs) at their C-termini and are primarily found at the plasma membrane, but they are also present in the nucleus and cytoplasm. ARF function and localization depends on their controlled binding and hydrolysis of GTP. Here we show that GTP-binding-defective ARL4D is targeted to the mitochondria, where it affects mitochondrial morphology and function. We found that a portion of endogenous ARL4D and the GTP-binding-defective ARL4D mutant ARL4D(T35N) reside in the mitochondria. The N-terminal myristoylation of ARL4D(T35N) was required for its localization to mitochondria. The localization of ARL4D(T35N) to the mitochondria reduced the mitochondrial membrane potential (ΔΨm) and caused mitochondrial fragmentation. Furthermore, the C-terminal NLS region of ARL4D(T35N) was required for its effect on the mitochondria. This study is the first to demonstrate that the dysfunctional GTP-binding-defective ARL4D is targeted to mitochondria, where it subsequently alters mitochondrial morphology and membrane potential.
Collapse
Affiliation(s)
- Chun-Chun Li
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tsung-Sheng Wu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Fang Huang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Li-Ting Jang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Tsan Liu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shu-Ting You
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Gunn-Guang Liou
- Division of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Fang-Jen S. Lee
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
28
|
Abstract
As plant Golgi bodies move through the cell along the actin cytoskeleton, they face the need to maintain their polarized stack structure whilst receiving, processing and distributing protein cargo destined for secretion. Structural proteins, or Golgi matrix proteins, help to hold cisternae together and tethering factors direct cargo carriers to the correct target membranes. This review focuses on golgins, a protein family containing long coiled-coil regions, summarizes their known functions in animal cells and highlights recent findings about plant golgins and their putative roles in the plant secretory pathway.
Collapse
Affiliation(s)
- A Osterrieder
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, UK.
| |
Collapse
|
29
|
Houghton FJ, Bellingham SA, Hill AF, Bourges D, Ang DK, Gemetzis T, Gasnereau I, Gleeson PA. Arl5b is a Golgi-localised small G protein involved in the regulation of retrograde transport. Exp Cell Res 2012; 318:464-77. [DOI: 10.1016/j.yexcr.2011.12.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 12/04/2011] [Accepted: 12/28/2011] [Indexed: 11/30/2022]
|
30
|
Direct imaging of RAB27B-enriched secretory vesicle biogenesis in lacrimal acinar cells reveals origins on a nascent vesicle budding site. PLoS One 2012; 7:e31789. [PMID: 22363735 PMCID: PMC3282733 DOI: 10.1371/journal.pone.0031789] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 01/16/2012] [Indexed: 12/20/2022] Open
Abstract
This study uses YFP-tagged Rab27b expression in rabbit lacrimal gland acinar cells, which are polarized secretory epithelial cells, to characterize early stages of secretory vesicle trafficking. Here we demonstrate the utility of YFP-Rab27b to delineate new perspectives on the mechanisms of early vesicle biogenesis in lacrimal gland acinar cells, where information is significantly limited. Protocols were developed to deplete the mature YFP-Rab27b-enriched secretory vesicle pool in the subapical region of the cell, and confocal fluorescence microscopy was used to track vesicle replenishment. This analysis revealed a basally-localized organelle, which we termed the "nascent vesicle site," from which nascent vesicles appeared to emerge. Subapical vesicular YFP-Rab27b was co-localized with p150(Glued), a component of the dynactin cofactor of cytoplasmic dynein. Treatment with the microtubule-targeted agent, nocodazole, did not affect release of mature secretory vesicles, although during vesicle repletion it significantly altered nascent YFP-Rab27b-enriched secretory vesicle localization. Instead of moving to the subapical region, these vesicles were trapped at the nascent vesicle site which was adjacent to, if not a sub-compartment of, the trans-Golgi network. Finally, YFP-Rab27b-enriched secretory vesicles which reached the subapical cytoplasm appeared to acquire the actin-based motor protein, Myosin 5C. Our findings show that Rab27b enrichment occurs early in secretory vesicle formation, that secretory vesicles bud from a visually discernable nascent vesicle site, and that transport from the nascent vesicle site to the subapical region requires intact microtubules.
Collapse
|
31
|
Abstract
Protein traffic is necessary to maintain homeostasis in all eukaryotic organisms. All newly synthesized secretory proteins destined to the secretory and endolysosmal systems are transported from the endoplasmic reticulum to the Golgi before delivery to their final destinations. Here, we describe the COPII and COPI coating machineries that generate carrier vesicles and the tethers and SNAREs that mediate COPII and COPI vesicle fusion at the ER-Golgi interface.
Collapse
Affiliation(s)
- Tomasz Szul
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
32
|
Christis C, Munro S. The small G protein Arl1 directs the trans-Golgi-specific targeting of the Arf1 exchange factors BIG1 and BIG2. ACTA ACUST UNITED AC 2012; 196:327-35. [PMID: 22291037 PMCID: PMC3275380 DOI: 10.1083/jcb.201107115] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Specificity in Arf1 GEF recruitment to the trans-Golgi, and thus in localized Arf1 activation, is provided by an Arf-like G protein. The small G protein Arf1 regulates Golgi traffic and is activated by two related types of guanine nucleotide exchange factor (GEF). GBF1 acts at the cis-Golgi, whereas BIG1 and its close paralog BIG2 act at the trans-Golgi. Peripheral membrane proteins such as these GEFs are often recruited to membranes by small G proteins, but the basis for specific recruitment of Arf GEFs, and hence Arfs, to Golgi membranes is not understood. In this paper, we report a liposome-based affinity purification method to identify effectors for small G proteins of the Arf family. We validate this with the Drosophila melanogaster Arf1 orthologue (Arf79F) and the related class II Arf (Arf102F), which showed a similar pattern of effector binding. Applying the method to the Arf-like G protein Arl1, we found that it binds directly to Sec71, the Drosophila ortholog of BIG1 and BIG2, via an N-terminal region. We show that in mammalian cells, Arl1 is necessary for Golgi recruitment of BIG1 and BIG2 but not GBF1. Thus, Arl1 acts to direct a trans-Golgi–specific Arf1 GEF, and hence active Arf1, to the trans side of the Golgi.
Collapse
Affiliation(s)
- Chantal Christis
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| | | |
Collapse
|
33
|
Hatakeyama H, Kanzaki M. Molecular basis of insulin-responsive GLUT4 trafficking systems revealed by single molecule imaging. Traffic 2011; 12:1805-20. [PMID: 21910807 DOI: 10.1111/j.1600-0854.2011.01279.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Development of a 'static retention' property of GLUT4, the insulin-responsive glucose transporter, has emerged as being essential for achieving its maximal insulin-induced surface exposure. Herein, employing quantum-dot-based nanometrology of intracellular GLUT4 behavior, we reveal the molecular basis of its systematization endowed upon adipogenic differentiation of 3T3L1 cells. Specifically, (i) the endosomes-to-trans-Golgi network (TGN) retrieval system specialized for GLUT4 develops in response to sortilin expression, which requires an intricately balanced interplay among retromers, golgin-97 and syntaxin-6, the housekeeping vesicle trafficking machinery. (ii) The Golgin-97-localizing subdomain of the differentiated TGN apparently serves as an intermediate transit route by which GLUT4 can further proceed to the stationary GLUT4 storage compartment. (iii) AS160/Tbc1d4 then renders the 'static retention' property insulin responsive, i.e. insulin liberates GLUT4 from the static state only in the presence of functional AS160/Tbc1d4. (iv) Moreover, sortilin malfunction and the resulting GLUT4 sorting defects along with retarded TGN function might be etiologically related to insulin resistance. Together, these observations provide a conceptual framework for understanding maturation/retardation of the insulin-responsive GLUT4 trafficking system that relies on the specialized subdomain of differentiated TGN.
Collapse
Affiliation(s)
- Hiroyasu Hatakeyama
- Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | | |
Collapse
|
34
|
Brown FC, Schindelhaim CH, Pfeffer SR. GCC185 plays independent roles in Golgi structure maintenance and AP-1-mediated vesicle tethering. ACTA ACUST UNITED AC 2011; 194:779-87. [PMID: 21875948 PMCID: PMC3171126 DOI: 10.1083/jcb.201104019] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
GCC185 is a long coiled-coil protein localized to the trans-Golgi network (TGN) that functions in maintaining Golgi structure and tethering mannose 6-phosphate receptor (MPR)-containing transport vesicles en route to the Golgi. We report the identification of two distinct domains of GCC185 needed either for Golgi structure maintenance or transport vesicle tethering, demonstrating the independence of these two functions. The domain needed for vesicle tethering binds to the clathrin adaptor AP-1, and cells depleted of GCC185 accumulate MPRs in transport vesicles that are AP-1 decorated. This study supports a previously proposed role of AP-1 in retrograde transport of MPRs from late endosomes to the Golgi and indicates that docking may involve the interaction of vesicle-associated AP-1 protein with the TGN-associated tethering protein GCC185.
Collapse
Affiliation(s)
- Frank C Brown
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | |
Collapse
|
35
|
Benjamin JJR, Poon PP, Drysdale JD, Wang X, Singer RA, Johnston GC. Dysregulated Arl1, a regulator of post-Golgi vesicle tethering, can inhibit endosomal transport and cell proliferation in yeast. Mol Biol Cell 2011; 22:2337-47. [PMID: 21562219 PMCID: PMC3128535 DOI: 10.1091/mbc.e10-09-0765] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Small monomeric G proteins regulated in part by GTPase-activating proteins (GAPs) are molecular switches for several aspects of vesicular transport. The yeast Gcs1 protein is a dual-specificity GAP for ADP-ribosylation factor (Arf) and Arf-like (Arl)1 G proteins, and also has GAP-independent activities. The absence of Gcs1 imposes cold sensitivity for growth and endosomal transport; here we present evidence that dysregulated Arl1 may cause these impairments. We show that gene deletions affecting the Arl1 or Ypt6 vesicle-tethering pathways prevent Arl1 activation and membrane localization, and restore growth and trafficking in the absence of Gcs1. A mutant version of Gcs1 deficient for both ArfGAP and Arl1GAP activity in vitro still allows growth and endosomal transport, suggesting that the function of Gcs1 that is required for these processes is independent of GAP activity. We propose that, in the absence of this GAP-independent regulation by Gcs1, the resulting dysregulated Arl1 prevents growth and impairs endosomal transport at low temperatures. In cells with dysregulated Arl1, an increased abundance of the Arl1 effector Imh1 restores growth and trafficking, and does so through Arl1 binding. Protein sequestration at the trans-Golgi membrane by dysregulated, active Arl1 may therefore be the mechanism of inhibition.
Collapse
Affiliation(s)
- Jeremy J R Benjamin
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 1X5
| | | | | | | | | | | |
Collapse
|
36
|
Chia PZC, Gleeson PA. The Regulation of Endosome-to-Golgi Retrograde Transport by Tethers and Scaffolds. Traffic 2011; 12:939-47. [DOI: 10.1111/j.1600-0854.2011.01185.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
37
|
Abstract
The trans-Golgi network (TGN) receives a select set of proteins from the endocytic pathway-about 5% of total plasma membrane glycoproteins (Duncan and Kornfeld 1988). Proteins that are delivered include mannose 6-phosphate receptors (MPRs), TGN46, sortilin, and various toxins that hitchhike a ride backward through the secretory pathway to intoxicate cells after they exit into the cytoplasm from the endoplasmic reticulum (ER). This article will review work on the molecular players that drive protein transport from the endocytic pathway to the TGN. Distinct requirements have revealed multiple routes for retrograde transport; in addition, the existence of multiple, potential coat proteins and/or cargo adaptors imply that multiple vesicular transfers are likely involved. Several comprehensive reviews have appeared recently and should be sought for additional details (Bonifacino and Rojas 2006; Johannes and Popoff 2008).
Collapse
Affiliation(s)
- Suzanne R Pfeffer
- Department of Biochemistry, Stanford University School of Medicine, California 94305-5307, USA.
| |
Collapse
|
38
|
TGN golgins, Rabs and cytoskeleton: regulating the Golgi trafficking highways. Trends Cell Biol 2010; 20:329-36. [DOI: 10.1016/j.tcb.2010.02.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2009] [Revised: 02/15/2010] [Accepted: 02/17/2010] [Indexed: 12/22/2022]
|
39
|
Sztul E, Lupashin V. Role of vesicle tethering factors in the ER-Golgi membrane traffic. FEBS Lett 2009; 583:3770-83. [PMID: 19887069 DOI: 10.1016/j.febslet.2009.10.083] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Revised: 10/29/2009] [Accepted: 10/30/2009] [Indexed: 12/27/2022]
Abstract
Tethers are a diverse group of loosely related proteins and protein complexes grouped into three families based on structural and functional similarities. A well-accepted role for tethering factors is the initial attachment of transport carriers to acceptor membranes prior to fusion. However, accumulating evidence indicates that tethers are more than static bridges. Tethers have been shown to interact with components of the fusion machinery and with components involved in vesicle formation. Tethers belonging to the three families act at the same stage of traffic, suggesting that they mediate distinct events during vesicle tethering. Thus, multiple tether-facilitated events are required to provide selectivity to vesicle fusion. In this review, we highlight findings that support this model.
Collapse
Affiliation(s)
- Elizabeth Sztul
- Department of Cell Biology, University of Alabama at Birmingham, 1918 University Boulevard, Birmingham, AL 35294, USA
| | | |
Collapse
|
40
|
The Localization of the Golgin GCC185 Is Independent of Rab6A/A' and Arl1. Cell 2009; 138:787-94. [DOI: 10.1016/j.cell.2009.05.048] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Revised: 03/14/2009] [Accepted: 05/26/2009] [Indexed: 12/21/2022]
|
41
|
Gage MC, Keen JN, Buxton AT, Bedi MK, Findlay JBC. Proteomic Analysis of IgE-Mediated Secretion by LAD2 Mast Cells. J Proteome Res 2009; 8:4116-25. [DOI: 10.1021/pr900108w] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Matthew C. Gage
- Institute of Membrane and Systems Biology, Faculty of Biological Sciences, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom, and Division of Cardiovascular and Diabetes Research, Faculty of Medicine and Health, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Jeffrey N. Keen
- Institute of Membrane and Systems Biology, Faculty of Biological Sciences, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom, and Division of Cardiovascular and Diabetes Research, Faculty of Medicine and Health, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Anthony T. Buxton
- Institute of Membrane and Systems Biology, Faculty of Biological Sciences, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom, and Division of Cardiovascular and Diabetes Research, Faculty of Medicine and Health, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Maninder K. Bedi
- Institute of Membrane and Systems Biology, Faculty of Biological Sciences, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom, and Division of Cardiovascular and Diabetes Research, Faculty of Medicine and Health, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - John B. C. Findlay
- Institute of Membrane and Systems Biology, Faculty of Biological Sciences, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom, and Division of Cardiovascular and Diabetes Research, Faculty of Medicine and Health, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
42
|
Nishimoto-Morita K, Shin HW, Mitsuhashi H, Kitamura M, Zhang Q, Johannes L, Nakayama K. Differential effects of depletion of ARL1 and ARFRP1 on membrane trafficking between the trans-Golgi network and endosomes. J Biol Chem 2009; 284:10583-92. [PMID: 19224922 DOI: 10.1074/jbc.m900847200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ARFRP1 and ARL1, which are both ARF-like small GTPases, are mammalian orthologs of yeast Arl3p and Arl1p, respectively. In yeast, Arl3p targeted to trans-Golgi network (TGN) membranes activates Arl1p, and the activated Arl1p in turn recruits a GRIP domain-containing protein; this complex regulates retrograde transport to the TGN and anterograde transport from the TGN. In the present study, using RNA interference-mediated knockdown of ARFRP1 and ARL1, we have examined whether the orthologs of Arl3p-Arl1p-GRIP story serve similar functions in mammalian cells. However, we have unexpectedly found differential roles of ARL1 and ARFRP1. Specifically, ARL1 and ARFRP1 regulate retrograde transport of Shiga toxin to the TGN and anterograde transport of VSVG from the TGN, respectively. Furthermore, we have obtained evidence suggesting that a SNARE complex containing Vti1a, syntaxin 6, and syntaxin 16 is involved in Shiga toxin transport downstream of ARL1.
Collapse
Affiliation(s)
- Kirika Nishimoto-Morita
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | | | | | |
Collapse
|
43
|
Hayes GL, Brown FC, Haas AK, Nottingham RM, Barr FA, Pfeffer SR. Multiple Rab GTPase binding sites in GCC185 suggest a model for vesicle tethering at the trans-Golgi. Mol Biol Cell 2009; 20:209-17. [PMID: 18946081 PMCID: PMC2613123 DOI: 10.1091/mbc.e08-07-0740] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 09/22/2008] [Accepted: 10/14/2008] [Indexed: 12/18/2022] Open
Abstract
GCC185, a trans-Golgi network-localized protein predicted to assume a long, coiled-coil structure, is required for Rab9-dependent recycling of mannose 6-phosphate receptors (MPRs) to the Golgi and for microtubule nucleation at the Golgi via CLASP proteins. GCC185 localizes to the Golgi by cooperative interaction with Rab6 and Arl1 GTPases at adjacent sites near its C terminus. We show here by yeast two-hybrid and direct biochemical tests that GCC185 contains at least four additional binding sites for as many as 14 different Rab GTPases across its entire length. A central coiled-coil domain contains a specific Rab9 binding site, and functional assays indicate that this domain is important for MPR recycling to the Golgi complex. N-Terminal coiled-coils are also required for GCC185 function as determined by plasmid rescue after GCC185 depletion by using small interfering RNA in cultured cells. Golgi-Rab binding sites may permit GCC185 to contribute to stacking and lateral interactions of Golgi cisternae as well as help it function as a vesicle tether.
Collapse
Affiliation(s)
- Garret L. Hayes
- *Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305; and
| | - Frank C. Brown
- *Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305; and
| | - Alexander K. Haas
- Cancer Research Centre, University of Liverpool, Liverpool L9 3AT, United Kingdom
| | - Ryan M. Nottingham
- *Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305; and
| | - Francis A. Barr
- Cancer Research Centre, University of Liverpool, Liverpool L9 3AT, United Kingdom
| | - Suzanne R. Pfeffer
- *Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305; and
| |
Collapse
|
44
|
Abstract
By presenting antigenic peptides on the cell surface, human leukocyte antigen (HLA) class I molecules are critical for immune defense. Their surface density determines, to a large extent, the level of CD8(+) T cell-dependent immune reactions; their loss is a major mechanism of immune escape. Therefore, powerful processes should regulate their surface expression. Here we document the mechanisms used by CD99 to mediate HLA class I modulation. Up-regulation of HLA class I by IFN-gamma requires CD99. In the trans Golgi network (TGN), and up to the cell surface, CD99 and HLA class I are physically associated via their transmembrane domain. CD99 also binds p230/golgin-245, a coiled-coil protein that recycles between the cytosol and buds/vesicles of the TGN and which plays a fundamental role in trafficking transport vesicles. p230/golgin-245 is anchored within TGN membranes via its Golgin-97, RanBP1, IMh1p, P230 (GRIP) domain and the overexpression of which leads to surface and intracellular down-modulation of HLA class I molecules.
Collapse
|
45
|
AtGRIP protein locates to the secretory vesicles of trans Golgi-network in Arabidopsis root cap cells. Sci Bull (Beijing) 2008. [DOI: 10.1007/s11434-008-0420-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
46
|
Guerriero CJ, Lai Y, Weisz OA. Differential sorting and Golgi export requirements for raft-associated and raft-independent apical proteins along the biosynthetic pathway. J Biol Chem 2008; 283:18040-7. [PMID: 18434305 PMCID: PMC2440606 DOI: 10.1074/jbc.m802048200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2008] [Revised: 04/16/2008] [Indexed: 12/29/2022] Open
Abstract
Sorting signals for apically destined proteins are highly diverse and can be present within the luminal, membrane-associated, and cytoplasmic domains of these proteins. A subset of apical proteins partition into detergent-resistant membranes, and the association of these proteins with glycolipid-enriched microdomains or lipid rafts may be important for their proper targeting. Recently, we observed that raft-associated and raft-independent apical proteins take different routes to the apical surface of polarized Madin-Darby canine kidney cells (Cresawn, K. O., Potter, B. A., Oztan, A., Guerriero, C. J., Ihrke, G., Goldenring, J. R., Apodaca, G., and Weisz, O. A. (2007) EMBO J. 26, 3737-3748). Here we reconstituted in vitro the export of raft-associated and raft-independent markers staged intracellularly at 19 degrees C. Surprisingly, whereas release of the raft-associated protein influenza hemagglutinin was dependent on the addition of an ATP-regenerating system and cytosol, release of a yellow fluorescent protein (YFP)-tagged raft-independent protein (the 75-kDa neurotrophin receptor; YFP-p75) was efficient even in the absence of these constituents. Subsequent studies suggested that YFP-p75 is released from the trans-Golgi network in fragile tubules that do not withstand isolation procedures. Moreover, immunofluorescence analysis revealed that hemagglutinin and YFP-p75 segregate into distinct subdomains of the Golgi complex at 19 degrees C. Our data suggest that raft-associated and raft-independent proteins accumulate at distinct intracellular sites upon low temperature staging, and that upon warming, they exit these compartments in transport carriers that have very different membrane characteristics and morphologies.
Collapse
Affiliation(s)
- Christopher J Guerriero
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
47
|
|
48
|
Burguete AS, Fenn TD, Brunger AT, Pfeffer SR. Rab and Arl GTPase family members cooperate in the localization of the golgin GCC185. Cell 2008; 132:286-98. [PMID: 18243103 DOI: 10.1016/j.cell.2007.11.048] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Revised: 10/31/2007] [Accepted: 11/26/2007] [Indexed: 10/22/2022]
Abstract
GCC185 is a large coiled-coil protein at the trans Golgi network that is required for receipt of transport vesicles inbound from late endosomes and for anchoring noncentrosomal microtubules that emanate from the Golgi. Here, we demonstrate that recruitment of GCC185 to the Golgi is mediated by two Golgi-localized small GTPases of the Rab and Arl families. GCC185 binds Rab6, and mutation of residues needed for Rab binding abolishes Golgi localization. The crystal structure of Rab6 bound to the GCC185 Rab-binding domain reveals that Rab6 recognizes a two-fold symmetric surface on a coiled coil immediately adjacent to a C-terminal GRIP domain. Unexpectedly, Rab6 binding promotes association of Arl1 with the GRIP domain. We present a structure-derived model for dual GTPase membrane attachment that highlights the potential ability of Rab GTPases to reach binding partners at a significant distance from the membrane via their unstructured and membrane-anchored, hypervariable domains.
Collapse
|
49
|
Abstract
Small G proteins play a central role in the organization of the secretory and endocytic pathways. The majority of such small G proteins are members of the Rab family, which are anchored to the bilayer by C-terminal prenyl groups. However, the recruitment of some effectors, including vesicle coat proteins, is mediated by a second class of small G proteins that is unique in having an N-terminal amphipathic helix that becomes available for membrane insertion upon GTP binding. Sar1, Arf1, and Arf6 are the best-characterized members of this ADP-ribosylation factor (Arf) family. In addition, all eukaryotes contain additional distantly related G proteins, often called Arf like, or Arls. The complete Arf family in humans has 29 members. The roles of these related G proteins are poorly understood, but recent work has shown that some are involved in membrane traffic or organizing the cytoskeleton. Here we review what is known about all the members of the Arf family, along with the known regulatory molecules that convert them between GDP- and GTP-bound states.
Collapse
|
50
|
Szul T, Grabski R, Lyons S, Morohashi Y, Shestopal S, Lowe M, Sztul E. Dissecting the role of the ARF guanine nucleotide exchange factor GBF1 in Golgi biogenesis and protein trafficking. J Cell Sci 2007; 120:3929-40. [PMID: 17956946 DOI: 10.1242/jcs.010769] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
COPI recruitment to membranes appears to be essential for the biogenesis of the Golgi and for secretory trafficking. Preventing COPI recruitment by expressing inactive forms of the ADP-ribosylation factor (ARF) or the ARF-activating guanine nucleotide exchange factor GBF1, or by treating cells with brefeldin A (BFA), causes the collapse of the Golgi into the endoplasmic reticulum (ER) and arrests trafficking of soluble and transmembrane proteins at the ER. Here, we assess COPI function in Golgi biogenesis and protein trafficking by preventing COPI recruitment to membranes by removing GBF1. We report that siRNA-mediated depletion of GBF1 causes COPI dispersal but does not lead to collapse of the Golgi. Instead, it causes extensive tubulation of the cis-Golgi. The Golgi-derived tubules target to peripheral ER-Golgi intermediate compartment (ERGIC) sites and create dynamic continuities between the ERGIC and the cis-Golgi compartment. COPI dispersal in GBF1-depleted cells causes dramatic inhibition of the trafficking of transmembrane proteins. Unexpectedly, soluble proteins continue to be secreted from GBF1-depleted cells. Our findings suggest that a secretory pathway capable of trafficking soluble proteins can be maintained in cells in which COPI recruitment is compromised by GBF1 depletion. However, the trafficking of transmembrane proteins through the existing pathway requires GBF1-mediated ARF activation and COPI recruitment.
Collapse
Affiliation(s)
- Tomasz Szul
- Department of Cell Biology, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL 35924, USA
| | - Robert Grabski
- Department of Cell Biology, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL 35924, USA
| | - Susan Lyons
- Department of Cell Biology, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL 35924, USA
| | - Yuichi Morohashi
- University of Manchester, Faculty of Life Sciences, Manchester M13 9PT, UK
| | - Svetlana Shestopal
- Department of Cell Biology, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL 35924, USA
| | - Martin Lowe
- University of Manchester, Faculty of Life Sciences, Manchester M13 9PT, UK
| | - Elizabeth Sztul
- Department of Cell Biology, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, AL 35924, USA
| |
Collapse
|