1
|
Schott MB, Rozeveld CN, Bhatt S, Crossman B, Krueger EW, Weller SG, Rasineni K, Casey CA, McNiven MA. Ethanol disrupts hepatocellular lipophagy by altering Rab5-centric LD-lysosome trafficking. Hepatol Commun 2024; 8:e0446. [PMID: 38780316 PMCID: PMC11124685 DOI: 10.1097/hc9.0000000000000446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/23/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Previous reports suggest that lipid droplets (LDs) in the hepatocyte can be catabolized by a direct engulfment from nearby endolysosomes (microlipophagy). Further, it is likely that this process is compromised by chronic ethanol (EtOH) exposure leading to hepatic steatosis. This study investigates the hepatocellular machinery supporting microlipophagy and EtOH-induced alterations in this process with a focus on the small, endosome-associated, GTPase Rab5. METHODS AND RESULTS Here we report that this small Ras-related GTPase is a resident component of LDs, and its activity is important for hepatocellular LD-lysosome proximity and physical interactions. We find that Rab5 siRNA knockdown causes an accumulation of LDs in hepatocytes by inhibiting lysosome dependent LD catabolism. Importantly, Rab5 appears to support this process by mediating the recruitment of early endosomal and or multivesicular body compartments to the LD surface before lysosome fusion. Interestingly, while wild-type or a constituently active GTPase form (Q79L) of Rab5 supports LD-lysosome transport, this process is markedly reduced in cells expressing a GTPase dead (S34N) Rab5 protein or in hepatocytes exposed to chronic EtOH. CONCLUSIONS These findings support the novel premise of an early endosomal/multivesicular body intermediate compartment on the LD surface that provides a "docking" site for lysosomal trafficking, not unlike the process that occurs during the hepatocellular degradation of endocytosed ligands that is also known to be compromised by EtOH exposure.
Collapse
Affiliation(s)
- Micah B. Schott
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Cody N. Rozeveld
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Saumya Bhatt
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Bridget Crossman
- Department of Biochemistry and Molecular Biology, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Eugene W. Krueger
- Department of Biochemistry and Molecular Biology, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Shaun G. Weller
- Department of Biochemistry and Molecular Biology, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Karuna Rasineni
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Veterans’ Affairs, VA-Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | - Carol A. Casey
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Veterans’ Affairs, VA-Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | - Mark A. McNiven
- Department of Biochemistry and Molecular Biology, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
2
|
Qiao L, Dong C, Jia W, Ma B. NAA20 recruits Rin2 and promotes triple-negative breast cancer progression by regulating Rab5A-mediated activation of EGFR signaling. Cell Signal 2023; 112:110922. [PMID: 37827343 DOI: 10.1016/j.cellsig.2023.110922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/25/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype with poor prognosis and high mortality. To improve the prognosis and survival of TNBC patients, it is necessary to explore new targets and signaling pathways to develop novel therapies for TNBC treatment. N-α-acetyltransferase 20 (NAA20) is one of the catalytic subunits of N-terminal acetyltransferase (NatB). It has been reported that NAA20 played a critical role in cancer progression. In this study, we found that NAA20 expression was markedly higher in TNBC tissues than in paracancerous normal tissues using The Cancer Genome Atlas (TCGA) analysis. This result was further confirmed by qRT-PCR and immunohistochemistry (IHC). Knockdown of NAA20 significantly inhibited TNBC cell viability by CCK8 and colony formation assays and cell migration and invasion by Transwell assays. Additionally, NAA20 knockdown decreased the expression of EGFR in TNBC cells. Upon stimulation with EGF and knockdown of NAA20, EGFR internalization and degradation were observed by confocal microscopy. The western blot results showed that NAA20 knockdown down-regulated PI3K, AKT, and mTOR phosphorylation. Next, we further explored the underlying molecular mechanisms of NAA20 by co-immunoprecipitation (Co-IP). The results suggested that there was an interacting relationship between NAA20 and Rab5A. Over-expression of NAA20 could potentiate the expression of Rab5A. Furthermore, the knockdown of Rab5A inhibited EGFR expression and the phosphorylation of downstream signaling targets. NAA20 over-expression offset the knockdown effect of Rab5A and activated EGFR signaling. Finally, we constructed a xenograft mouse model transfected TNBC cells to investigate the role of NAA20 in vivo. NAA20 knockdown markedly suppressed tumor growth and decreased tumor volume and weight. In conclusion, our study demonstrated that NAA20, a novel target of TNBC, could promote TNBC progression by regulating Rab5A-mediated activation of EGFR signaling.
Collapse
Affiliation(s)
- Lei Qiao
- Department of Breast and Thyroid Surgery, Xinjiang Medical University affiliated Tumor Hospital, Urumqi, Xinjiang Uygur Autonomous Region 830000, China
| | - Chao Dong
- Department of Breast and Thyroid Surgery, Xinjiang Medical University affiliated Tumor Hospital, Urumqi, Xinjiang Uygur Autonomous Region 830000, China
| | - Wenlei Jia
- Department of Breast and Thyroid Surgery, Xinjiang Medical University affiliated Tumor Hospital, Urumqi, Xinjiang Uygur Autonomous Region 830000, China
| | - Binlin Ma
- Department of Breast and Thyroid Surgery, Xinjiang Medical University affiliated Tumor Hospital, Urumqi, Xinjiang Uygur Autonomous Region 830000, China.
| |
Collapse
|
3
|
The Rab GTPase in the heart: Pivotal roles in development and disease. Life Sci 2022; 306:120806. [PMID: 35841978 DOI: 10.1016/j.lfs.2022.120806] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 07/03/2022] [Accepted: 07/11/2022] [Indexed: 11/20/2022]
Abstract
Rab proteins are a family of small GTPases that function as molecular switches of intracellular vesicle formation and membrane trafficking. As a key factor, Rab GTPase participates in autophagy and protein transport and acts as the central hub of membrane trafficking in eukaryotes. The role of Rab GTPase in neurodegenerative disorders, such as Alzheimer's and Parkinson's, has been extensively investigated; however, its implication in cardiovascular embryogenesis and diseases remains largely unknown. In this review, we summarize previous findings and reveal their importance in the onset and progression of cardiac diseases, as well as their emergence as potential therapeutic targets for cardiovascular disease.
Collapse
|
4
|
Hatoyama Y, Homma Y, Hiragi S, Fukuda M. Establishment and analysis of conditional Rab1- and Rab5-knockout cells using the auxin-inducible degron system. J Cell Sci 2021; 134:273782. [PMID: 34817057 DOI: 10.1242/jcs.259184] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/11/2021] [Indexed: 12/13/2022] Open
Abstract
Two small GTPases, Rab1 and Rab5, are key membrane trafficking regulators that are conserved in all eukaryotes. They have recently been found to be essential for cell survival and/or growth in cultured mammalian cells, thereby precluding the establishment of Rab1-knockout (KO) and Rab5-KO cells, making it extremely difficult to assess the impact of complete Rab1 or Rab5 protein depletion on cellular functions. Here, we generated and analyzed cell lines with conditional KO (CKO) of either Rab1 (Rab1A and Rab1B) or Rab5 (Rab5A, Rab5B and Rab5C) by using the auxin-inducible protein degradation system. Rab1 CKO and Rab5 CKO led to eventual cell death from 18 h and 48 h, respectively, after auxin exposure. After acute Rab1 protein depletion, the Golgi stack and ribbon structures were completely disrupted, and endoplasmic reticulum (ER)-to-Golgi trafficking was severely inhibited. Moreover, we discovered a novel Rab1-depletion phenotype: perinuclear clustering of early endosomes and delayed transferrin recycling. In contrast, acute Rab5 protein depletion resulted in loss of early endosomes and late endosomes, but lysosomes appeared to be normal. We also observed a dramatic reduction in the intracellular signals of endocytic cargos via receptor-mediated or fluid-phase endocytosis in Rab5-depleted cells.
Collapse
Affiliation(s)
- Yuki Hatoyama
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Yuta Homma
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Shu Hiragi
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
5
|
Trofimenko E, Homma Y, Fukuda M, Widmann C. The endocytic pathway taken by cationic substances requires Rab14 but not Rab5 and Rab7. Cell Rep 2021; 37:109945. [PMID: 34731620 DOI: 10.1016/j.celrep.2021.109945] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/23/2021] [Accepted: 10/13/2021] [Indexed: 02/01/2023] Open
Abstract
Endocytosis and endosome dynamics are controlled by proteins of the small GTPase Rab family. Besides possible recycling routes to the plasma membrane and various organelles, previously described endocytic pathways (e.g., clathrin-mediated endocytosis, macropinocytosis, CLIC/GEEC pathway) all appear to funnel the endocytosed material to Rab5-positive early endosomes that then mature into Rab7-positive late endosomes/lysosomes. By studying the uptake of a series of cell-penetrating peptides (CPPs), we identify an endocytic pathway that moves material to nonacidic Lamp1-positive late endosomes. Trafficking via this endocytic route is fully independent of Rab5 and Rab7 but requires the Rab14 protein. The pathway taken by CPPs differs from the conventional Rab5-dependent endocytosis at the stage of vesicle formation already, as it is not affected by a series of compounds that inhibit macropinocytosis or clathrin-mediated endocytosis. The Rab14-dependent pathway is also used by physiological cationic molecules such as polyamines and homeodomains found in homeoproteins.
Collapse
Affiliation(s)
- Evgeniya Trofimenko
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Yuta Homma
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Christian Widmann
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
6
|
Harms FL, Parthasarathy P, Zorndt D, Alawi M, Fuchs S, Halliday BJ, McKeown C, Sampaio H, Radhakrishnan N, Radhakrishnan SK, Gorce M, Navet B, Ziegler A, Sachdev R, Robertson SP, Nampoothiri S, Kutsche K. Biallelic loss-of-function variants in TBC1D2B cause a neurodevelopmental disorder with seizures and gingival overgrowth. Hum Mutat 2020; 41:1645-1661. [PMID: 32623794 DOI: 10.1002/humu.24071] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 06/08/2020] [Accepted: 06/30/2020] [Indexed: 12/15/2022]
Abstract
The family of Tre2-Bub2-Cdc16 (TBC)-domain containing GTPase activating proteins (RABGAPs) is not only known as key regulatorof RAB GTPase activity but also has GAP-independent functions. Rab GTPases are implicated in membrane trafficking pathways, such as vesicular trafficking. We report biallelic loss-of-function variants in TBC1D2B, encoding a member of the TBC/RABGAP family with yet unknown function, as the underlying cause of cognitive impairment, seizures, and/or gingival overgrowth in three individuals from unrelated families. TBC1D2B messenger RNA amount was drastically reduced, and the protein was absent in fibroblasts of two patients. In immunofluorescence analysis, ectopically expressed TBC1D2B colocalized with vesicles positive for RAB5, a small GTPase orchestrating early endocytic vesicle trafficking. In two independent TBC1D2B CRISPR/Cas9 knockout HeLa cell lines that serve as cellular model of TBC1D2B deficiency, epidermal growth factor internalization was significantly reduced compared with the parental HeLa cell line suggesting a role of TBC1D2B in early endocytosis. Serum deprivation of TBC1D2B-deficient HeLa cell lines caused a decrease in cell viability and an increase in apoptosis. Our data reveal that loss of TBC1D2B causes a neurodevelopmental disorder with gingival overgrowth, possibly by deficits in vesicle trafficking and/or cell survival.
Collapse
Affiliation(s)
- Frederike L Harms
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Padmini Parthasarathy
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Dennis Zorndt
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malik Alawi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sigrid Fuchs
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin J Halliday
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Colina McKeown
- Centre for Clinical Genetics, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Hugo Sampaio
- Department of Women and Children's Health, University of New South Wales, Randwick Campus, Randwick, NSW, Australia.,Sydney Children's Hospital, Randwick, NSW, Australia
| | - Natasha Radhakrishnan
- Department of Ophthalmology, Amrita Institute of Medical Sciences and Research Centre, Cochin, Kerala, India
| | - Suresh K Radhakrishnan
- Department of Neurology, Amrita Institute of Medical Sciences and Research Centre, Cochin, Kerala, India
| | - Magali Gorce
- Department of Metabolic Disease, Children University Hospital, Toulouse, France
| | - Benjamin Navet
- Department of Biochemistry and Genetics, University Hospital of Angers, Angers, France.,MitoLab, Institut MitoVasc, UMR CNRS6015, INSERM U1083, Angers, France
| | - Alban Ziegler
- Department of Biochemistry and Genetics, University Hospital of Angers, Angers, France.,MitoLab, Institut MitoVasc, UMR CNRS6015, INSERM U1083, Angers, France
| | - Rani Sachdev
- Centre for Clinical Genetics, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Stephen P Robertson
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Sheela Nampoothiri
- Department of Pediatric Genetics, Amrita Institute of Medical Sciences and Research Centre, Cochin, Kerala, India
| | - Kerstin Kutsche
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
7
|
Zhou X, Xie S, Wu S, Qi Y, Wang Z, Zhang H, Lu D, Wang X, Dong Y, Liu G, Yang D, Shi Q, Bian W, Yu R. Golgi phosphoprotein 3 promotes glioma progression via inhibiting Rab5-mediated endocytosis and degradation of epidermal growth factor receptor. Neuro Oncol 2018; 19:1628-1639. [PMID: 28575494 DOI: 10.1093/neuonc/nox104] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Golgi phosphoprotein 3 (GOLPH3) is associated with worse prognosis of gliomas, but its role and mechanism in glioma progression remain largely unknown. This study aimed to explore the role and mechanism of GOLPH3 in glioma progression. Methods The expression of GOLPH3 in glioma tissues was detected by quantitative PCR, immunoblotting, and immunohistochemistry. GOLPH3's effect on glioma progression was examined using cell growth assays and an intracranial glioma model. The effect of GOLPH3 on epidermal growth factor receptor (EGFR) stability, endocytosis, and degradation was examined by immunoblotting and immunofluorescence. The activity of Rab5 was checked by glutathione S-transferase pulldown assay. Results GOLPH3 was upregulated in gliomas, and its downregulation inhibited glioma cell proliferation both in vitro and in vivo. Furthermore, GOLPH3 depletion dampened EGFR signaling by enhancing EGFR endocytosis, driving EGFR into late endosome and promoting lysosome-mediated degradation. Interestingly, GOLPH3 bound to Rab5 and GOLPH3 downregulation promoted the activation of Rab5. In addition, Rab5 depletion abolished the effect of GOLPH3 on EGFR endocytosis and degradation. Conclusion Our results imply that GOLPH3 promotes glioma cell proliferation via inhibiting Rab5-mediated endocytosis and degradation of EGFR, thereby activating the phosphatidylinositol-3 kinase (PI3K)-Akt-mammalian target of rapamycin (mTOR) signaling pathway. We find a new mechanism by which GOLPH3 promotes tumor progression through regulating cell surface receptor trafficking. Extensive and intensive understanding of the role of GOLPH3 in glioma progression may provide an opportunity to develop a novel molecular therapeutic target for gliomas.
Collapse
Affiliation(s)
- Xiuping Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shao Xie
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shishuang Wu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanhua Qi
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhaohao Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hao Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dong Lu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xu Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu Dong
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Guanzheng Liu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dongxu Yang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qiong Shi
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wenbin Bian
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
8
|
Magnetic capture of polydopamine-encapsulated Hela cells for the analysis of cell surface proteins. J Proteomics 2018; 172:76-81. [DOI: 10.1016/j.jprot.2017.10.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 09/25/2017] [Accepted: 10/12/2017] [Indexed: 12/16/2022]
|
9
|
Murray M, Zhou F. Trafficking and other regulatory mechanisms for organic anion transporting polypeptides and organic anion transporters that modulate cellular drug and xenobiotic influx and that are dysregulated in disease. Br J Pharmacol 2017; 174:1908-1924. [PMID: 28299773 DOI: 10.1111/bph.13785] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/01/2017] [Accepted: 03/05/2017] [Indexed: 12/25/2022] Open
Abstract
Organic anion transporters (OATs) and organic anion-transporting polypeptides (OATPs), encoded by a number of solute carrier (SLC)22A and SLC organic anion (SLCO) genes, mediate the absorption and distribution of drugs and other xenobiotics. The regulation of OATs and OATPs is complex, comprising both transcriptional and post-translational mechanisms. Plasma membrane expression is required for cellular substrate influx by OATs/OATPs. Thus, interest in post-translational regulatory processes, including membrane targeting, endocytosis, recycling and degradation of transporter proteins, is increasing because these are critical for plasma membrane expression. After being synthesized, transporters undergo N-glycosylation in the endoplasmic reticulum and Golgi apparatus and are delivered to the plasma membrane by vesicular transport. Their expression at the cell surface is maintained by de novo synthesis and recycling, which occurs after clathrin- and/or caveolin-dependent endocytosis of existing protein. Several studies have shown that phosphorylation by signalling kinases is important for the internalization and recycling processes, although the transporter protein does not appear to be directly phosphorylated. After internalization, transporters that are targeted for degradation undergo ubiquitination, most likely on intracellular loop residues. Epigenetic mechanisms, including methylation of gene regulatory regions and transcription from alternate promoters, are also significant in the regulation of certain SLC22A/SLCO genes. The membrane expression of OATs/OATPs is dysregulated in disease, which affects drug efficacy and detoxification. Several transporters are expressed in the cytoplasmic subcompartment in disease states, which suggests that membrane targeting/internalization/recycling may be impaired. This article focuses on recent developments in OAT and OATP regulation, their dysregulation in disease and the significance for drug therapy.
Collapse
Affiliation(s)
- Michael Murray
- Pharmacogenomics and Drug Development Group, Discipline of Pharmacology, School of Medical Sciences, The University of Sydney, NSW, 2006, Australia
| | - Fanfan Zhou
- Faculty of Pharmacy, The University of Sydney, NSW, 2006, Australia
| |
Collapse
|
10
|
Ghosh P, Aznar N, Swanson L, Lo IC, Lopez-Sanchez I, Ear J, Rohena C, Kalogriopoulos N, Joosen L, Dunkel Y, Sun N, Nguyen P, Bhandari D. Biochemical, Biophysical and Cellular Techniques to Study the Guanine Nucleotide Exchange Factor, GIV/Girdin. CURRENT PROTOCOLS IN CHEMICAL BIOLOGY 2016; 8:265-298. [PMID: 27925669 PMCID: PMC5154557 DOI: 10.1002/cpch.13] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Canonical signal transduction via heterotrimeric G proteins is spatiotemporally restricted, i.e., triggered exclusively at the plasma membrane, only by agonist activation of G protein-coupled receptors via a finite process that is terminated within a few hundred milliseconds. Recently, a rapidly emerging paradigm has revealed a noncanonical pathway for activation of heterotrimeric G proteins via the nonreceptor guanidine-nucleotide exchange factor, GIV/Girdin. Biochemical, biophysical, and functional studies evaluating this pathway have unraveled its unique properties and distinctive spatiotemporal features. As in the case of any new pathway/paradigm, these studies first required an in-depth optimization of tools/techniques and protocols, governed by rationale and fundamentals unique to the pathway, and more specifically to the large multimodular GIV protein. Here we provide the most up-to-date overview of protocols that have generated most of what we know today about noncanonical G protein activation by GIV and its relevance in health and disease. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093-0651
- Department of Medicine, University of California at San Diego, La Jolla, CA 92093-0651
| | - Nicolas Aznar
- Department of Medicine, University of California at San Diego, La Jolla, CA 92093-0651
| | - Lee Swanson
- Department of Medicine, University of California at San Diego, La Jolla, CA 92093-0651
| | - I-Chung Lo
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093-0651
| | | | - Jason Ear
- Department of Medicine, University of California at San Diego, La Jolla, CA 92093-0651
| | - Cristina Rohena
- Department of Medicine, University of California at San Diego, La Jolla, CA 92093-0651
| | | | - Linda Joosen
- Department of Medicine, University of California at San Diego, La Jolla, CA 92093-0651
| | - Ying Dunkel
- Department of Medicine, University of California at San Diego, La Jolla, CA 92093-0651
| | - Nina Sun
- Department of Medicine, University of California at San Diego, La Jolla, CA 92093-0651
| | - Peter Nguyen
- Department of Chemistry and Biochemistry, California State University Long Beach, Long Beach, CA 90840-9507
| | - Deepali Bhandari
- Department of Chemistry and Biochemistry, California State University Long Beach, Long Beach, CA 90840-9507
| |
Collapse
|
11
|
Rush JS, Bingaman DP, Chaney PG, Wax MB, Ceresa BP. Administration of Menadione, Vitamin K3, Ameliorates Off-Target Effects on Corneal Epithelial Wound Healing Due to Receptor Tyrosine Kinase Inhibition. Invest Ophthalmol Vis Sci 2016; 57:5864-5871. [PMID: 27802516 PMCID: PMC6733502 DOI: 10.1167/iovs.16-19952] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 09/25/2016] [Indexed: 12/11/2022] Open
Abstract
PURPOSE The antiangiogenic receptor tyrosine kinase inhibitor (RTKi), 3-[(4-bromo-2,6-difluorophenyl)methoxy]-5-[[[[4-(1-pyrrolidinyl) butyl] amino] carbonyl]amino]-4-isothiazolecarboxamide hydrochloride, targets VEGFR2 (half maximal inhibitory concentration [IC50] = 11 nM); however, off-target inhibition of epidermal growth factor receptor (EGFR) occurs at higher concentrations. (IC50 = 5.8 μM). This study was designed to determine the effect of topical RTKi treatment on EGF-mediated corneal epithelial wound healing and to develop new strategies to minimize off-target EGFR inhibition. METHODS In vitro corneal epithelial wound healing was measured in response to EGF using a transformed human cell line (hTCEpi cells). In vivo corneal wound healing was assessed using a murine model. In these complementary assays, wound healing was measured in the presence of varying RTKi concentrations. Immunoblot analysis was used to examine EGFR and VEGFR2 phosphorylation and the kinetics of EGFR degradation. An Alamar Blue assay measured VEGFR2-mediated cell biology. RESULTS Receptor tyrosine kinase inhibitor exposure caused dose-dependent inhibition of EGFR-mediated corneal epithelial wound healing in vitro and in vivo. Nanomolar concentrations of menadione, a vitamin K3 analog, when coadministered with the RTKi, slowed EGFR degradation and ameliorated the inhibitory effects on epithelial wound healing both in vitro and in vivo. Menadione did not alter the RTKi's IC50 against VEGFR2 phosphorylation or its inhibition of VEGF-induced retinal endothelial cell proliferation. CONCLUSIONS An antiangiogenic RTKi exhibited off-target effects on the corneal epithelium that can be minimized by menadione without deleteriously affecting its on-target VEGFR2 blockade. These data indicate that menadione has potential as a topical supplement for individuals suffering from perturbations in corneal epithelial homeostasis, especially as an untoward side effect of kinase inhibitors.
Collapse
Affiliation(s)
- Jamie S Rush
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, United States
| | | | - Paul G Chaney
- PanOptica, Inc., Bernardsville, New Jersey, United States
| | - Martin B Wax
- PanOptica, Inc., Bernardsville, New Jersey, United States 3Department of Ophthalmology and Visual Sciences, Rutgers New Jersey Medical School Newark, New Jersey, United States
| | - Brian P Ceresa
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, United States 4Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, Kentucky, United States
| |
Collapse
|
12
|
CMTM7 knockdown increases tumorigenicity of human non-small cell lung cancer cells and EGFR-AKT signaling by reducing Rab5 activation. Oncotarget 2016; 6:41092-107. [PMID: 26528697 PMCID: PMC4747392 DOI: 10.18632/oncotarget.5732] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 09/19/2015] [Indexed: 12/20/2022] Open
Abstract
The dysregulation of epidermal growth factor receptor (EGFR) signaling has been well documented to contribute to the progression of non-small cell lung cancer (NSCLC), the leading cause of cancer death in the world. EGF-stimulated EGFR activation induces receptor internalization and degradation, which plays an important role in EGFR signaling. This process is frequently deregulated in cancer cells, leading to enhanced EGFR levels and signaling. Our previous study on CMTM7 is only limited to a brief description of the relationship of overexpressed CMTM7 with EGFR-AKT signaling. The biological functions of endogenous CMTM7 and its molecular mechanism remained unclear. In this study, we show that the stable knockdown of CMTM7 augments the malignant potential of NSCLC cells and enhances EGFR-AKT signaling by decreasing EGFR internalization and degradation. Mechanistically, CMTM7 knockdown reduces the activation of Rab5, a protein known to be required for early endosome fusion. In NSCLC, the loss of CMTM7 would therefore serve to sustain aberrant EGFR-mediated oncogenic signaling. Together, our findings highlight the role of CMTM7 in the regulation of EGFR signaling in tumor cells, revealing CMTM7 as a novel molecule related to Rab5 activation.
Collapse
|
13
|
Neves LF, Duan J, Voelker A, Khanal A, McNally L, Steinbach-Rankins J, Ceresa BP. Preparation and optimisation of anionic liposomes for delivery of small peptides and cDNA to human corneal epithelial cells. J Microencapsul 2016; 33:391-9. [PMID: 27530524 DOI: 10.1080/02652048.2016.1202343] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Drug delivery to corneal epithelial cells is challenging due to the intrinsic mechanisms that protect the eye. Here, we report a novel liposomal formulation to encapsulate and deliver a short sequence peptide into human corneal epithelial cells (hTCEpi). Using a mixture of Phosphatidylcholine/Caproylamine/Dioleoylphosphatidylethanolamine (PC/CAP/DOPE), we encapsulated a fluorescent peptide, resulting in anionic liposomes with an average size of 138.8 ± 34 nm and a charge of -18.2 ± 1.3 mV. After 2 h incubation with the peptide-encapsulated liposomes, 66% of corneal epithelial (hTCEpi) cells internalised the FITC-labelled peptide, demonstrating the ability of this formulation to effectively deliver peptide to hTCEpi cells. Additionally, lipoplexes (liposomes complexed with plasmid DNA) were also able to transfect hTCEpi cells, albeit at a modest level (8% of the cells). Here, we describe this novel anionic liposomal formulation intended to enhance the delivery of small cargo molecules in situ.
Collapse
Affiliation(s)
- Luís F Neves
- a Department of Pharmacology and Toxicology , University of Louisville , Louisville , KY , USA
| | - Jinghua Duan
- b Department of Bioengineering , University of Louisville , Louisville , KY , USA ;,c Centre for Predictive Medicine , University of Louisville , Louisville , KY , USA
| | - Adrienne Voelker
- a Department of Pharmacology and Toxicology , University of Louisville , Louisville , KY , USA
| | - Anil Khanal
- d Brown Cancer Centre , University of Louisville , Louisville , KY , USA
| | - Lacey McNally
- d Brown Cancer Centre , University of Louisville , Louisville , KY , USA
| | - Jill Steinbach-Rankins
- a Department of Pharmacology and Toxicology , University of Louisville , Louisville , KY , USA ;,b Department of Bioengineering , University of Louisville , Louisville , KY , USA ;,c Centre for Predictive Medicine , University of Louisville , Louisville , KY , USA
| | - Brian P Ceresa
- a Department of Pharmacology and Toxicology , University of Louisville , Louisville , KY , USA ;,d Brown Cancer Centre , University of Louisville , Louisville , KY , USA
| |
Collapse
|
14
|
Juillot S, Cott C, Madl J, Claudinon J, van der Velden NSJ, Künzler M, Thuenauer R, Römer W. Uptake of Marasmius oreades agglutinin disrupts integrin-dependent cell adhesion. Biochim Biophys Acta Gen Subj 2015; 1860:392-401. [PMID: 26546712 PMCID: PMC4717121 DOI: 10.1016/j.bbagen.2015.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 10/31/2015] [Accepted: 11/03/2015] [Indexed: 11/18/2022]
Abstract
BACKGROUND Fruiting body lectins have been proposed to act as effector proteins in the defense of fungi against parasites and predators. The Marasmius oreades agglutinin (MOA) is a lectin from the fairy ring mushroom with specificity for Galα1-3Gal containing carbohydrates. This lectin is composed of an N-terminal carbohydrate-binding domain and a C-terminal dimerization domain. The dimerization domain of MOA shows in addition calcium-dependent cysteine protease activity, similar to the calpain family. METHODS Cell detachment assay, cell viability assay, immunofluorescence, live cell imaging and Western blot using MDCKII cell line. RESULTS In this study, we demonstrate in MDCKII cells that after internalization, MOA protease activity induces profound physiological cellular responses, like cytoskeleton rearrangement, cell detachment and cell death. These changes are preceded by a decrease in FAK phosphorylation and an internalization and degradation of β1-integrin, consistent with a disruption of integrin-dependent cell adhesion signaling. Once internalized, MOA accumulates in late endosomal compartments. CONCLUSION Our results suggest a possible toxic mechanism of MOA, which consists of disturbing the cell adhesion and the cell viability. GENERAL SIGNIFICANCE After being ingested by a predator, MOA might exert a protective role by diminishing host cell integrity.
Collapse
Affiliation(s)
- Samuel Juillot
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Schänzlestraße 1, D-79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, D-79104 Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestraße 18, D-79104 Freiburg, Germany
| | - Catherine Cott
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Schänzlestraße 1, D-79104 Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestraße 18, D-79104 Freiburg, Germany
| | - Josef Madl
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Schänzlestraße 1, D-79104 Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestraße 18, D-79104 Freiburg, Germany
| | - Julie Claudinon
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Schänzlestraße 1, D-79104 Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestraße 18, D-79104 Freiburg, Germany
| | | | - Markus Künzler
- Institute of Microbiology, Department of Biology, ETH Zürich, Vladimir-Prelog-Weg 4, CH-8093 Zürich, Switzerland
| | - Roland Thuenauer
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Schänzlestraße 1, D-79104 Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestraße 18, D-79104 Freiburg, Germany
| | - Winfried Römer
- Faculty of Biology, Albert-Ludwigs-University Freiburg, Schänzlestraße 1, D-79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, D-79104 Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestraße 18, D-79104 Freiburg, Germany.
| |
Collapse
|
15
|
Chua CEL, Tang BL. Role of Rab GTPases and their interacting proteins in mediating metabolic signalling and regulation. Cell Mol Life Sci 2015; 72:2289-304. [PMID: 25690707 PMCID: PMC11113524 DOI: 10.1007/s00018-015-1862-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/09/2015] [Accepted: 02/11/2015] [Indexed: 10/24/2022]
Abstract
The vesicular transport pathways, which shuttle materials to and from the cell surface and within the cell, and the metabolic (growth factor and nutrient) signalling pathways, which integrate a variety of extracellular and intracellular signals to mediate growth, proliferation or survival, are both important for cellular physiology. There is evidence to suggest that the transport and metabolic signalling pathways intersect-vesicular transport can affect the regulation of metabolic signals and vice versa. The Rab family GTPases regulate the specificity of vesicular transport steps in the cell. Together with their interacting proteins, Rabs would likely constitute the points of intersection between vesicular transport and metabolic signalling pathways. Examples of these points would include growth factor signalling, glucose and lipid metabolism, as well as autophagy. Many of these processes involve mechanistic/mammalian target of rapamycin (mTOR) complex 1 (mTORC1) in downstream cascades, or are regulated by TORC signalling. A general functionality of the vesicular transport processes controlled by the Rabs is also important for spatial and temporal regulation of the transmission of metabolic signals between the cell surface and the nucleus. In other cases, specific Rabs and their interacting proteins are known to function in recruiting metabolism-related proteins to target membranes, or may compete with other factors in the TORC signalling pathway as a means of metabolic regulation. We review and discuss herein examples of how Rabs and their interacting proteins can mediate metabolic signalling and regulation in cells.
Collapse
Affiliation(s)
- Christelle En Lin Chua
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, 8 Medical Drive, Singapore, 117597, Singapore,
| | | |
Collapse
|
16
|
Hoepflinger MC, Geretschlaeger A, Sommer A, Hoeftberger M, Hametner C, Ueda T, Foissner I. Molecular Analysis and Localization of CaARA7 a Conventional RAB5 GTPase from Characean Algae. Traffic 2015; 16:534-54. [PMID: 25639563 PMCID: PMC4898595 DOI: 10.1111/tra.12267] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 01/27/2015] [Accepted: 01/27/2015] [Indexed: 11/28/2022]
Abstract
RAB5 GTPases are important regulators of endosomal membrane traffic. Among them Arabidopsis thaliana ARA7/RABF2b is highly conserved and homologues are present in fungal, animal and plant kingdoms. In land plants ARA7 and its homologues are involved in endocytosis and transport towards the vacuole. Here we report on the isolation of an ARA7 homologue (CaARA7/CaRABF2) in the highly evolved characean green alga Chara australis. It encodes a polypeptide of 202 amino acids with a calculated molecular mass of 22.2 kDa and intrinsic GTPase activity. Immunolabelling of internodal cells with a specific antibody reveals CaARA7 epitopes at multivesicular endosomes (MVEs) and at MVE-containing wortmannin (WM) compartments. When transiently expressed in epidermal cells of Nicotiana benthamiana leaves, fluorescently tagged CaARA7 localizes to small organelles (putative MVEs) and WM compartments, and partially colocalizes with AtARA7 and CaARA6, a plant specific RABF1 GTPase. Mutations in membrane anchoring and GTP binding sites alter localization of CaARA7 and affect GTPase activity, respectively. This first detailed study of a conventional RAB5 GTPase in green algae demonstrates that CaARA7 is similar to RAB5 GTPases from land plants and other organisms and shows conserved structure and localization.
Collapse
Affiliation(s)
- Marion C. Hoepflinger
- Department of Cell Biology/Plant Physiology, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | - Anja Geretschlaeger
- Department of Cell Biology/Plant Physiology, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | - Aniela Sommer
- Department of Cell Biology/Plant Physiology, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | - Margit Hoeftberger
- Department of Cell Biology/Plant Physiology, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | - Christina Hametner
- Department of Organismic Biology, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | - Takashi Ueda
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Ilse Foissner
- Department of Cell Biology/Plant Physiology, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| |
Collapse
|
17
|
Studzian M, Bartosz G, Pulaski L. Endocytosis of ABCG2 drug transporter caused by binding of 5D3 antibody: trafficking mechanisms and intracellular fate. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1759-71. [PMID: 25918011 DOI: 10.1016/j.bbamcr.2015.04.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 03/31/2015] [Accepted: 04/14/2015] [Indexed: 12/16/2022]
Abstract
ABCG2, a metabolite and xenobiotic transporter located at the plasma membrane (predominantly in barrier tissues and progenitor cells), undergoes a direct progressive endocytosis process from plasma membrane to intracellular compartments upon binding of 5D3 monoclonal antibody. This antibody is specific to an external epitope on the protein molecule and locks it in a discrete conformation within its activity cycle, presumably providing a structural trigger for the observed internalization phenomenon. Using routine and novel assays, we show that ABCG2 is endocytosed by a mixed mechanism: partially via a rapid, clathrin-dependent pathway and partially in a cholesterol-dependent, caveolin-independent manner. While the internalization process is entirely dynamin-dependent and converges initially at the early endosome, subsequent intracellular fate of ABCG2 is again twofold: endocytosis leads to only partial lysosomal degradation, while a significant fraction of the protein is retained in a post-endosomal compartment with the possibility of at least partial recycling back to the cell surface. This externally triggered, conformation-related trafficking pathway may serve as a general regulatory paradigm for membrane transporters, and its discovery was made possible thanks to consistent application of quantitative methods.
Collapse
Affiliation(s)
- Maciej Studzian
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., 90-237 Lodz, Poland
| | - Grzegorz Bartosz
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., 90-237 Lodz, Poland
| | - Lukasz Pulaski
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., 90-237 Lodz, Poland; Laboratory of Transcriptional Regulation, Institute of Medical Biology PAS, Lodowa 106, 93-232 Lodz, Poland.
| |
Collapse
|
18
|
Hudson SV, Huang JS, Yin W, Albeituni S, Rush J, Khanal A, Yan J, Ceresa BP, Frieboes HB, McNally LR. Targeted noninvasive imaging of EGFR-expressing orthotopic pancreatic cancer using multispectral optoacoustic tomography. Cancer Res 2014; 74:6271-9. [PMID: 25217521 DOI: 10.1158/0008-5472.can-14-1656] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Detection of orthotopic xenograft tumors is difficult due to poor spatial resolution and reduced image fidelity with traditional optical imaging modalities. In particular, light scattering and attenuation in tissue at depths beyond subcutaneous implantation hinder adequate visualization. We evaluate the use of multispectral optoacoustic tomography (MSOT) to detect upregulated epidermal growth factor (EGF) receptor in orthotopic pancreatic xenografts using a near-infrared EGF-conjugated CF-750 fluorescent probe. MSOT is based on the photoacoustic effect and thus not limited by photon scattering, resulting in high-resolution tomographic images. Pancreatic tumor-bearing mice with luciferase-transduced S2VP10L tumors were intravenously injected with EGF-750 probe before MSOT imaging. We characterized probe specificity and bioactivity via immunoblotting, immunocytochemistry, and flow cytometric analysis. In vitro data along with optical bioluminescence/fluorescence imaging were used to validate acquired MSOT in vivo images of probe biodistribution. Indocyanine green dye was used as a nonspecific control to define specificity of EGF-probe accumulation. Maximum accumulation occurred at 6 hours postinjection, demonstrating specific intratumoral probe uptake and minimal liver and kidney off-target accumulation. Optical bioluminescence and fluorescence imaging confirmed tumor-specific probe accumulation consistent with MSOT images. These studies demonstrate the utility of MSOT to obtain volumetric images of ligand probe biodistribution in vivo to detect orthotopic pancreatic tumor lesions through active targeting of the EGF receptor.
Collapse
Affiliation(s)
| | - Justin S Huang
- Department of Medicine, University of Louisville, Kentucky
| | - Wenyuan Yin
- Department of Medicine, University of Louisville, Kentucky
| | | | - Jamie Rush
- Department of Pharmacology and Toxicology, University of Louisville, Kentucky
| | - Anil Khanal
- Department of Medicine, University of Louisville, Kentucky
| | - Jun Yan
- Department of Medicine, University of Louisville, Kentucky
| | - Brian P Ceresa
- Department of Pharmacology and Toxicology, University of Louisville, Kentucky
| | - Hermann B Frieboes
- Department of Pharmacology and Toxicology, University of Louisville, Kentucky. Department of Bioengineering, University of Louisville, Kentucky
| | - Lacey R McNally
- Department of Medicine, University of Louisville, Kentucky. Department of Pharmacology and Toxicology, University of Louisville, Kentucky.
| |
Collapse
|
19
|
Kinsey C, Balakrishnan V, O'Dell MR, Huang JL, Newman L, Whitney-Miller CL, Hezel AF, Land H. Plac8 links oncogenic mutations to regulation of autophagy and is critical to pancreatic cancer progression. Cell Rep 2014; 7:1143-55. [PMID: 24794439 DOI: 10.1016/j.celrep.2014.03.061] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 02/12/2014] [Accepted: 03/24/2014] [Indexed: 12/20/2022] Open
Abstract
Mutations in p53 and RAS potently cooperate in oncogenic transformation, and correspondingly, these genetic alterations frequently coexist in pancreatic ductal adenocarcinoma (PDA) and other human cancers. Previously, we identified a set of genes synergistically activated by combined RAS and p53 mutations as frequent downstream mediators of tumorigenesis. Here, we show that the synergistically activated gene Plac8 is critical for pancreatic cancer growth. Silencing of Plac8 in cell lines suppresses tumor formation by blocking autophagy, a process essential for maintaining metabolic homeostasis in PDA, and genetic inactivation in an engineered mouse model inhibits PDA progression. We show that Plac8 is a critical regulator of the autophagic machinery, localizing to the lysosomal compartment and facilitating lysosome-autophagosome fusion. Plac8 thus provides a mechanistic link between primary oncogenic mutations and the induction of autophagy, a central mechanism of metabolic reprogramming, during PDA progression.
Collapse
Affiliation(s)
- Conan Kinsey
- Department of Biomedical Genetics, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Vijaya Balakrishnan
- Department of Biomedical Genetics, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Michael R O'Dell
- Department of Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Jing Li Huang
- Department of Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Laurel Newman
- Department of Biomedical Genetics, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Christa L Whitney-Miller
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Aram F Hezel
- Department of Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA; James P. Wilmot Cancer Center, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| | - Hartmut Land
- Department of Biomedical Genetics, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA; James P. Wilmot Cancer Center, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| |
Collapse
|
20
|
Chua CEL, Tang BL. Engagement of the small GTPase Rab31 protein and its effector, early endosome antigen 1, is important for trafficking of the ligand-bound epidermal growth factor receptor from the early to the late endosome. J Biol Chem 2014; 289:12375-89. [PMID: 24644286 DOI: 10.1074/jbc.m114.548321] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Rab31 is a member of the Rab5 subfamily of Rab GTPases. Although localized largely to the trans-Golgi network, it shares common guanine nucleotide exchange factors and effectors with other Rab5 subfamily members that have been implicated in endocytic membrane traffic. We investigated whether Rab31 also has a role in the trafficking of the ligand-bound EGF receptor (EGFR) internalized through receptor-mediated endocytosis. We found that loss of Rab31 inhibits, but overexpression enhances, EGFR trafficking to the late endosomes and that the effect of Rab31 silencing could be specifically rescued by overexpression of a silencing-resistant form of Rab31. Rab31 was found to interact with the EGFR by coimmunoprecipitation and affinity pulldown analyses, and the primarily trans-Golgi network-localized Rab31 has increased colocalization with the EGFR in A431 cells 30 min after pulsing with EGF. A glycerol gradient sedimentation assay suggested that Rab31 is sequestered into a high molecular weight complex after stimulation with EGF, as was early endosome antigen 1 (EEA1), a factor responsible for endosomal tethering and fusion events. We found that loss of EEA1 reduced the interaction between Rab31 and the EGFR and abrogated the effect of Rab31 overexpression on the trafficking of the EGFR. Likewise, loss of GAPex5, a Rab31 guanine nucleotide exchange factor that has a role in ubiquitination and degradation of the EGFR, reduced the interaction of Rab31 with the EGFR and its effect on EGFR trafficking. Taken together, our results suggest that Rab31 is an important regulator of endocytic trafficking of the EGFR and functions in an EGFR trafficking complex that includes EEA1 and GAPex5.
Collapse
Affiliation(s)
- Christelle En Lin Chua
- From the Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, 1E Kent Ridge Road, Singapore 119228 and
| | | |
Collapse
|
21
|
Rush JS, Ceresa BP. RAB7 and TSG101 are required for the constitutive recycling of unliganded EGFRs via distinct mechanisms. Mol Cell Endocrinol 2013; 381:188-97. [PMID: 23933150 PMCID: PMC3831653 DOI: 10.1016/j.mce.2013.07.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 07/26/2013] [Accepted: 07/31/2013] [Indexed: 12/13/2022]
Abstract
Both constitutive and ligand-mediated membrane trafficking regulate Epidermal Growth Factor Receptor (EGFR) signaling. The constitutive endocytosis and recycling of the unliganded EGFR is a critical determinant of cell surface EGFR expression and the cell's sensitivity to ligands. We report that two proteins with established roles in trafficking the EGF:EGFR complex to the lysosome also regulate the recycling of the unliganded EGFR. Knock down of either Tumor suppressor gene 101 (TSG101) or RAB7 causes the endosomal accumulation of the inactive, unliganded receptor in morphologically and biochemically distinct organelles. Knock down of TSG101 causes the EGFR to accumulate in low density endosomes whereas RAB7 knock down results in EGFR accumulation in high density endosomes. Knock down of either protein caused the receptor to co-localize primarily with LAMP-1, but not EEA1. These two proteins regulate EGFR slow, perinuclear recycling, via distinct mechanism and are new molecular targets that regulate cell surface EGFR expression.
Collapse
Affiliation(s)
- Jamie S. Rush
- Department of Pharmacology and Toxicology, University of Louisville
| | - Brian P. Ceresa
- Department of Pharmacology and Toxicology, University of Louisville
- Department of Cell Biology, University of Oklahoma Health Sciences Center
| |
Collapse
|
22
|
Johnston DA, Tapia AL, Eberle KE, Palmer GE. Three prevacuolar compartment Rab GTPases impact Candida albicans hyphal growth. EUKARYOTIC CELL 2013; 12:1039-50. [PMID: 23709183 PMCID: PMC3697461 DOI: 10.1128/ec.00359-12] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 05/20/2013] [Indexed: 11/20/2022]
Abstract
Disruption of vacuolar biogenesis in the pathogenic yeast Candida albicans causes profound defects in polarized hyphal growth. However, the precise vacuolar pathways involved in yeast-hypha differentiation have not been determined. Previously we focused on Vps21p, a Rab GTPase involved in directing vacuolar trafficking through the late endosomal prevacuolar compartment (PVC). Herein, we identify two additional Vps21p-related GTPases, Ypt52p and Ypt53p, that colocalize with Vps21p and can suppress the hyphal defects of the vps21Δ/Δ mutant. Phenotypic analysis of gene deletion strains revealed that loss of both VPS21 and YPT52 causes synthetic defects in endocytic trafficking to the vacuole, as well as delivery of the virulence-associated vacuolar membrane protein Mlt1p from the Golgi compartment. Transcription of all three GTPase-encoding genes is increased under hyphal growth conditions, and overexpression of the transcription factor Ume6p is sufficient to increase the transcription of these genes. While only the vps21Δ/Δ single mutant has hyphal growth defects, these were greatly exacerbated in a vps21Δ/Δ ypt52Δ/Δ double mutant. On the basis of relative expression levels and phenotypic analysis of gene deletion strains, Vps21p is the most important of the three GTPases, followed by Ypt52p, while Ypt53p has an only marginal impact on C. albicans physiology. Finally, disruption of a nonendosomal AP-3-dependent vacuolar trafficking pathway in the vps21Δ/Δ ypt52Δ/Δ mutant, further exacerbated the stress and hyphal growth defects. These findings underscore the importance of membrane trafficking through the PVC in sustaining the invasive hyphal growth form of C. albicans.
Collapse
Affiliation(s)
- Douglas A Johnston
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, School of Medicine, New Orleans, Louisiana, USA
| | | | | | | |
Collapse
|
23
|
Ceresa BP. Spatial regulation of epidermal growth factor receptor signaling by endocytosis. Int J Mol Sci 2012; 14:72-87. [PMID: 23344022 DOI: 10.3390/ijms14010072] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 12/10/2012] [Accepted: 12/12/2012] [Indexed: 12/26/2022] Open
Abstract
Signaling by cell surface receptors appears to be relatively straight-forward: ligand binds to the extracellular domain of the receptor and biochemical changes are communicated into the cell. However, this process is more complex than it first seems due to the various mechanisms that regulate signaling. In order to effectively target these receptors for pharmacological purposes, a more complete understanding of how their signaling is regulated is needed. Here, how the endocytic pathway regulates receptor signaling is discussed, using the epidermal growth factor receptor (EGFR) as a model. In particular, the spatial regulation of signaling is examined. Areas of discussion include: how endocytic trafficking affects biology/pathology, varying approaches for studying the relationship between receptor endocytosis and signaling, and developments in how the endocytic pathway controls EGFR:effector communication and EGFR-mediated cell biology.
Collapse
Affiliation(s)
- Brian P Ceresa
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
24
|
Stokes L. Rab5 regulates internalisation of P2X4 receptors and potentiation by ivermectin. Purinergic Signal 2012; 9:113-21. [PMID: 23086000 DOI: 10.1007/s11302-012-9336-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 10/08/2012] [Indexed: 01/21/2023] Open
Abstract
The P2X4 receptor is an ATP-gated ion channel expressed in neurons, endothelia and immune cells. Plasma membrane expression of P2X4 is regulated by dynamin-dependent endocytosis, and this study identifies a Rab5-dependent pathway of receptor internalisation. Expression of Rab5 constructs altered the distribution of P2X4 in HEK-293 cells, and both constitutive internalisation and agonist-induced desensitisation of P2X4 were increased by co-expression of wild-type Rab5 or constitutively active Rab5 (Q79L). Expression of inactive dynamin K44A and Rab5 S34N constructs abolished agonist-induced desensitisation, suggesting internalisation as the underlying mechanism. Blocking P2X4 internalisation in this way also abolished potentiation of ATP-induced currents by the allosteric modulator ivermectin. This suggests that the dynamin-Rab5 internalisation pathway is essential for the ivermectin potentiation effect. In agreement with this hypothesis, the co-expression of wild-type dynamin, wild-type Rab5 or active Rab5 (Q79L) could increase the potentiation of the ATP-induced P2X4 response by ivermectin. These findings highlight Rab5 GTPase as a key regulator of P2X4 receptor cell surface expression and internalisation.
Collapse
Affiliation(s)
- Leanne Stokes
- Department of Biomedical Science, University of Sheffield, Sheffield, UK.
| |
Collapse
|
25
|
Pedersen NM, Raiborg C, Brech A, Skarpen E, Roxrud I, Platta HW, Liestøl K, Stenmark H. The PtdIns3P-binding protein Phafin 2 mediates epidermal growth factor receptor degradation by promoting endosome fusion. Traffic 2012; 13:1547-63. [PMID: 22816767 DOI: 10.1111/j.1600-0854.2012.01400.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 07/17/2012] [Accepted: 07/20/2012] [Indexed: 12/16/2022]
Abstract
Phosphatidylinositol 3-phosphate (PtdIns3P) orchestrates endosomal cargo transport, fusion and motility by recruiting FYVE or PX domain-containing effector proteins to endosomal membranes. In an attempt to discover novel PtdIns3P effectors involved in the termination of growth factor receptor signalling, we performed an siRNA screen for epidermal growth factor (EGF) degradation, targeting FYVE and PX domain proteins in the human proteome. This screen identified several potential regulators of EGF degradation, including HRS (used as positive control), PX kinase, MTMR4 and Phafin2/PLEKHF2. As Phafin2 has not previously been shown to be required for EGF receptor (EGFR) degradation, we performed further functional studies on this protein. Loss of Phafin2 was found to decrease early endosome size, whereas overexpression of Phafin2 resulted in enlarged endosomes. Moreover, both the EGFR and the fluid-phase marker dextran were retained in abnormally small endosomes in Phafin2-depleted cells. In yeast two-hybrid analysis we identified Phafin2 as a novel interactor of the endosomal-tethering protein EEA1, and Phafin2 colocalized strongly with EEA1 in microdomains of the endosome membrane. Our results suggest that Phafin2 controls receptor trafficking and fluid-phase transport through early endosomes by facilitating endosome fusion in concert with EEA1.
Collapse
Affiliation(s)
- Nina Marie Pedersen
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, N-0310, Oslo, Norway
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Rush JS, Quinalty LM, Engelman L, Sherry DM, Ceresa BP. Endosomal accumulation of the activated epidermal growth factor receptor (EGFR) induces apoptosis. J Biol Chem 2011; 287:712-722. [PMID: 22102283 DOI: 10.1074/jbc.m111.294470] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Endocytosis positively and negatively regulates cell surface receptor signaling by temporally and spatially controlling interactions with downstream effectors. This process controls receptor-effector communication. However, the relationship between receptor endocytic trafficking and cell physiology is unclear. In MDA-MB-468 cells, cell surface EGF receptors (EGFRs) promote cell growth, whereas intracellular EGFRs induce apoptosis, making these cells an excellent model for studying the endocytic regulation of EGFR signaling. In addition, MDA-MB-468 cells have limited EGFR degradation following stimulation. Here, we report that in MDA-MB-468 cells the phosphorylated EGFR accumulates on the limiting membrane of the endosome with its carboxyl terminus oriented to the cytoplasm. To determine whether perturbation of EGFR trafficking is sufficient to cause apoptosis, we used pharmacological and biochemical strategies to disrupt EGFR endocytic trafficking in HeLa cells, which do not undergo EGF-dependent apoptosis. Manipulation of HeLa cells so that active EGF·EGFRs accumulate on the limiting membrane of endosomes reveals that receptor phosphorylation is sustained and leads to apoptosis. When EGF·EGFR complexes accumulated in the intraluminal vesicles of the late endosome, phosphorylation of the receptor was not sustained, nor did the cells undergo apoptosis. These data demonstrate that EGFR-mediated apoptosis is initiated by the activated EGFR from the limiting membrane of the endosome.
Collapse
Affiliation(s)
- Jamie S Rush
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73106
| | - Leslie M Quinalty
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73106
| | - Luke Engelman
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73106
| | - David M Sherry
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73106; Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73106; Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73106
| | - Brian P Ceresa
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73106.
| |
Collapse
|
27
|
Coon BG, Crist S, González-Bonet AM, Kim HK, Sowa J, Thompson DH, Ratliff TL, Aguilar RC. Fibronectin attachment protein from bacillus Calmette-Guerin as targeting agent for bladder tumor cells. Int J Cancer 2011; 131:591-600. [PMID: 21901746 DOI: 10.1002/ijc.26413] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 08/10/2011] [Indexed: 12/13/2022]
Abstract
The adjuvant therapy of choice for superficial bladder cancer is the intravesical instillation of live Mycobacterium bovis bacillus Calmette-Guerin (BCG). Despite the fact that this therapy is the most effective treatment for superficial bladder cancer, intravesical administration of BCG is associated with high local morbidity and the potential for systemic infection. Therefore, there is a need for the development of safer, less toxic approaches to fight this disease. Because fibronectin attachment protein (FAP) is a key element in BCG retention and targeting to cells, we hypothesize that this protein can be used as targeting agent to deliver cytotoxic cargo for the treatment of bladder tumors. Here, we evaluated the ability of bladder tumor cells to bind and endocytose FAP via fibronectin-integrin complexes. We found that microaggregation induced by an anti-FAP polyclonal antibody accelerated FAP uptake by T24 bladder tumor cells. FAP was determined to be internalized via a clathrin-independent, caveolae-dependent mechanism. Furthermore, once within the endosomal compartment, FAP was targeted to the lysosomal compartment with negligible recycling to the plasma membrane. Importantly, we demonstrated that FAP microaggregation and internalization could also be triggered by multivalent Ni(2+) NTA-bearing liposomes. Overall, our studies validate the use of FAP as a targeting vector and provide the foundation for the design of more effective, less-toxic bladder cancer therapeutics.
Collapse
Affiliation(s)
- Brian G Coon
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Lee JR, Hahn HS, Kim YH, Nguyen HH, Yang JM, Kang JS, Hahn MJ. Adaptor protein containing PH domain, PTB domain and leucine zipper (APPL1) regulates the protein level of EGFR by modulating its trafficking. Biochem Biophys Res Commun 2011; 415:206-11. [PMID: 22037462 DOI: 10.1016/j.bbrc.2011.10.064] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Accepted: 10/12/2011] [Indexed: 02/04/2023]
Abstract
The EGFR-mediated signaling pathway regulates multiple biological processes such as cell proliferation, survival and differentiation. Previously APPL1 (adaptor protein containing PH domain, PTB domain and leucine zipper 1) has been reported to function as a downstream effector of EGF-initiated signaling. Here we demonstrate that APPL1 regulates EGFR protein levels in response to EGF stimulation. Overexpression of APPL1 enhances EGFR stabilization while APPL1 depletion by siRNA reduces EGFR protein levels. APPL1 depletion accelerates EGFR internalization and movement of EGF/EGFR from cell surface to the perinuclear region in response to EGF treatment. Conversely, overexpression of APPL1 decelerates EGFR internalization and translocation of EGF/EGFR to the perinuclear region. Furthermore, APPL1 depletion enhances the activity of Rab5 which is involved in internalization and trafficking of EGFR and inhibition of Rab5 in APPL1-depleted cells restored EGFR levels. Consistently, APPL1 depletion reduced activation of Akt, the downstream signaling effector of EGFR and this is restored by inhibition of Rab5. These findings suggest that APPL1 is required for EGFR signaling by regulation of EGFR stabilities through inhibition of Rab5.
Collapse
Affiliation(s)
- Jae-Rin Lee
- Department of Molecular Cell Biology, Center for Molecular Medicine, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
29
|
Huang TH, Ka SM, Hsu YJ, Shui HA, Tang BL, Hu KY, Chang JL, Chen A. Rab23 plays a role in the pathophysiology of mesangial cells--a proteomic analysis. Proteomics 2011; 11:380-94. [PMID: 21268268 DOI: 10.1002/pmic.201000165] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2010] [Revised: 10/13/2010] [Accepted: 11/02/2010] [Indexed: 12/23/2022]
Abstract
Rab23, a novel member of the Rab family of small GTPases, has recently been identified in mesangial cells (MCs). Although Rab23 levels in MCs are associated with glomerular nephropathies, the exact physiological and pathological roles of Rab23 in MCs are unknown. In the present study, its roles in MCs were explored by performing proteomics and systems biology analyses in MCs after knockdown or overexpression of Rab23. Knockdown of Rab23 was achieved by transfecting MCs with a plasmid expressing short hairpin RNA against Rab23, while overexpression of Rab23 was accomplished by transfection with the wild-type, dominant negative, and constitutively active Rab23 gene constructs. The effects of different levels of Rab23 activity on proteome of various biological pathways were investigated. Gel-based proteomic approaches and systems biology tools, respectively, were used to identify the Rab23-regulated proteins and the functional pathways. Proteomic analysis revealed the potential roles for Rab23 in multiple processes, including G-protein signal transduction, transcription modulation, RNA stabilization, protein synthesis and degradation, cytoskeleton reorganization, anti-oxidation and detoxification, circadian rhythm regulation and phagocytosis. Bioinformatics analyses showed that Rab23 impacts on multiple biological networks in MCs. These data may shed light on the roles of Rab23 in mesangiopathy or MC damage.
Collapse
Affiliation(s)
- Tzu-Hao Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
30
|
West MB, Hanigan MH. γ-Glutamyl transpeptidase is a heavily N-glycosylated heterodimer in HepG2 cells. Arch Biochem Biophys 2010; 504:177-81. [PMID: 20831856 DOI: 10.1016/j.abb.2010.08.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Revised: 08/24/2010] [Accepted: 08/29/2010] [Indexed: 01/02/2023]
Abstract
The cell surface enzyme γ-glutamyl transpeptidase (GGT) is expressed by human hepatocellular carcinomas (HCCs). HCCs arise from malignant transformation of hepatocytes and are the most common form of primary liver cancer. Identification of tumor-specific, post-translational modifications of GGT may provide novel biomarkers for HCC. The HepG2 cell line, derived from a human HCC, has been used extensively in studies of liver cancer. However, the use of this cell line for studies of GGT have been stymied by reports that HepG2 cells do not process the GGT propeptide into its heterodimeric subunits. The data in this study demonstrate that HepG2 cells do, in fact, produce the mature heterodimeric form of GGT. Immunohistochemical and immunoaffinity analyses provide direct evidence that, in HepG2 cells, GGT is properly localized to the bile canaliculi. Three independent, experimental approaches demonstrate that GGT in HepG2 cells is comprised of two subunits that are more heavily N-glycosylated than GGT from normal human liver tissue. These data directly contradict the dogma in the field. These data support the use of HepG2 cells as a model system for analyzing tumor-specific changes in the post-translational modifications of GGT.
Collapse
Affiliation(s)
- Matthew B West
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, 73104, USA
| | | |
Collapse
|
31
|
Matsuda JJ, Filali MS, Moreland JG, Miller FJ, Lamb FS. Activation of swelling-activated chloride current by tumor necrosis factor-alpha requires ClC-3-dependent endosomal reactive oxygen production. J Biol Chem 2010; 285:22864-73. [PMID: 20479003 DOI: 10.1074/jbc.m109.099838] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
ClC-3 is a Cl(-)/H(+) antiporter required for cytokine-induced intraendosomal reactive oxygen species (ROS) generation by Nox1. ClC-3 current is distinct from the swelling-activated chloride current (ICl(swell)), but overexpression of ClC-3 can activate currents that resemble ICl(swell). Because H(2)O(2) activates ICl(swell) directly, we hypothesized that ClC-3-dependent, endosomal ROS production activates ICl(swell). Whole-cell perforated patch clamp methods were used to record Cl(-) currents in cultured aortic vascular smooth muscle cells from wild type (WT) and ClC-3 null mice. Under isotonic conditions, tumor necrosis factor-alpha (TNF-alpha) (10 ng/ml) activated outwardly rectifying Cl(-) currents with time-dependent inactivation in WT but not ClC-3 null cells. Inhibition by tamoxifen (10 microm) and by hypertonicity (340 mosm) identified them as ICl(swell). ICl(swell) was also activated by H(2)O(2) (500 microm), and the effect of TNF-alpha was completely inhibited by polyethylene glycol-catalase. ClC-3 expression induced ICl(swell) in ClC-3 null cells in the absence of swelling or TNF-alpha, and this effect was also blocked by catalase. ICl(swell) activation by hypotonicity (240 mosm) was only partially inhibited by catalase, and the size of these currents did not differ between WT and ClC-3 null cells. Disruption of endosome trafficking with either mutant Rab5 (S34N) or Rab11 (S25N) inhibited TNF-alpha-mediated activation of ICl(swell). Thrombin also activates ROS production by Nox1 but not in endosomes. Thrombin caused H(2)O(2)-dependent activation of ICl(swell), but this effect was not ClC-3- or Rab5-dependent. Thus, activation of ICl(swell) by TNF-alpha requires ClC-3-dependent endosomal H(2)O(2) production. This demonstrates a functional link between two distinct anion currents, ClC-3 and ICl(swell).
Collapse
Affiliation(s)
- James J Matsuda
- Department of Pediatrics, University of Iowa Children's Hospital and the University of Iowa Hospitals and Clinics, Iowa City, Iowa 52242, USA
| | | | | | | | | |
Collapse
|
32
|
McClintock JL, Ceresa BP. Transforming growth factor-{alpha} enhances corneal epithelial cell migration by promoting EGFR recycling. Invest Ophthalmol Vis Sci 2010; 51:3455-61. [PMID: 20181835 DOI: 10.1167/iovs.09-4386] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
PURPOSE. The goal of this study was to determine the molecular mechanism by which transforming growth factor-alpha (TGF-alpha) is a more potent activator of epidermal growth factor receptor (EGFR)-mediated corneal wound healing than epidermal growth factor (EGF). METHODS. Telomerase immortalized human corneal epithelial (hTCEpi) cells and primary human corneal epithelial cells were tested for their ability to migrate in response to EGF and TGF-alpha. In parallel, the endocytic trafficking of the EGFR in response to these same ligands was examined using indirect immunofluorescence, immunoblots, and radioligand binding. RESULTS. TGF-alpha, compared with EGF, is a more potent activator of corneal epithelial cell migration. Although both TGF-alpha and EGF were able to induce EGFR internalization and phosphorylation, only those receptors that were stimulated with EGF progressed to lysosomal degradation. EGFRs stimulated with TGF-alpha recycled back to the plasma membrane, where they could be reactivated with ligand. CONCLUSIONS. This study reveals that EGFR-mediated cell migration is limited by ligand-stimulated downregulation of the EGFR. This limitation can be overcome by treating cells with TGF-alpha because TGF-alpha stimulates EGFR endocytosis, but not degradation. After internalization of the TGF-alpha/EGFR complex, EGFR recycles back to the plasma membrane, where it can be restimulated. This sequence of events provides the receptor multiple opportunities for stimulation. Thus, stimulation with TGF-alpha prolongs EGFR signaling compared with EGF.
Collapse
Affiliation(s)
- Jennifer L McClintock
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | | |
Collapse
|
33
|
Peng M, Yin N, Zhang W. Endocytosis of FcalphaR is clathrin and dynamin dependent, but its cytoplasmic domain is not required. Cell Res 2009; 20:223-37. [PMID: 19859085 DOI: 10.1038/cr.2009.120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
FcalphaR, the Fc receptor for IgA, is essential for IgA-mediated immune responses. Previous studies have shown that IgA and IgA immune complexes can be rapidly endocytosed by FcalphaR. However, the underlying mechanism remains unclear. Here, we investigated the endocytic pathway of FcalphaR in monocytic cell line, U937, that naturally express FcalphaR and in transfected Chinese hamster ovary (CHO), COS-7 and Hela cells. By using selective chemical inhibitors of different endocytic pathways, overexpression of dominant-negative mutants of Eps15 and knockdown of clathrin heavy chain (CHC) via RNA interference, we demonstrated that endocytosis of FcalphaR was through a clathrin-mediated pathway. The endocytosed FcalphaR went into Rab5- and Rab11-positive endosomes. However, endocytosis of FcalphaR could not be blocked by a dominant-negative mutant of Rab5. We also demonstrated that endocytosis of FcalphaR was dynamin-dependent by overexpressing a dominant-negative mutant of dynamin. The potential endocytic motif for FcalphaR was also examined. Unexpectedly, we found that the entire cytoplasmic domain of FcalphaR was not required for the endocytic process of FcalphaR. We conclude that endocytosis of FcalphaR is clathrin- and dynamin-dependent, but is not regulated by Rab5, and the endocytic motif is not located in the cytoplasmic domain of FcalphaR.
Collapse
Affiliation(s)
- Min Peng
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | | | | |
Collapse
|
34
|
Wegner CS, Wegener CS, Malerød L, Pedersen NM, Progida C, Prodiga C, Bakke O, Stenmark H, Brech A. Ultrastructural characterization of giant endosomes induced by GTPase-deficient Rab5. Histochem Cell Biol 2009; 133:41-55. [PMID: 19830447 DOI: 10.1007/s00418-009-0643-8] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2009] [Indexed: 02/02/2023]
Abstract
The small GTPase Rab5 controls the fusogenic properties of early endosomes through GTP-dependent recruitment and activation of effector proteins. Expression of a GTPase-defective mutant, Rab5(Q79L), is known to cause formation of enlarged early endosomes. The ability of Rab5-GTP to recruit multiple effectors raises the question whether the Rab5(Q79L)-induced giant endosomes simply represent enlarged early endosomes or whether they have a more complex phenotype. In this report, we have addressed this issue by generating a HEp2 cell line with inducible expression of Rab5(Q79L) and performing ultrastructural analysis of Rab5(Q79L)-induced endosomes. We find that Rab5(Q79L) not only induces formation of enlarged early endosomes but also causes enlargement of later endocytic profiles. Most strikingly, Rab5(Q79L) causes formation of enlarged multivesicular endosomes with a large number of intraluminal vesicles, and endosomes that contain both early and late endocytic markers are frequently observed. In addition, we observe defects in the sorting of the EGF receptor and the transferrin receptor through this compartment.
Collapse
Affiliation(s)
- Catherine Sem Wegner
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, Oslo, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Chen PI, Kong C, Su X, Stahl PD. Rab5 isoforms differentially regulate the trafficking and degradation of epidermal growth factor receptors. J Biol Chem 2009; 284:30328-38. [PMID: 19723633 DOI: 10.1074/jbc.m109.034546] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ligand-mediated endocytosis is an intricate regulatory mechanism for epidermal growth factor receptor (EGFR) signal transduction. Coordinated trafficking of EGFR ensures its temporal and spatial communication with downstream signaling effectors. We focused our work on Rab5, a monomeric GTPase shown to participate in early stages of the endocytic pathway. Rab5 has three isoforms (A, B, and C) sharing more than 90% of sequence identity. We individually ablated endogenous isoforms in HeLa cells with short interfering RNAs and examined the loss-of-function phenotypes. We found that suppression of Rab5A or 5B hampered the degradation of EGFR, whereas Rab5C depletion had very little effect. The differential delay of EGFR degradation also corresponds with retarded progression of EGFR from early to late endosomes. We investigated the activators/effectors of Rab5A that can potentially separate its potency in EGFR degradation from other isoforms and found that Rin1, a Rab5 exchange factor, preferably associated with Rab5A. Moreover, Rab5A activation is sensitive to EGF stimulation, and suppression of Rin1 diminished this sensitivity. Based on our results together with previous work showing that Rin1 interacts with signal transducing adapter molecule to facilitate the degradation of EGFR (Kong, C., Su, X., Chen, P. I., and Stahl, P. D. (2007) J. Biol. Chem. 282, 15294-15301), we hypothesize that the selective association of Rab5A and Rin1 contributes to the dominance of Rab5A in EGFR trafficking, whereas the other isoforms may have major functions unrelated to the EGFR degradation pathway.
Collapse
Affiliation(s)
- Pin-I Chen
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
36
|
Jopling HM, Odell A, Hooper NM, Zachary IC, Walker JH, Ponnambalam S. Rab GTPase regulation of VEGFR2 trafficking and signaling in endothelial cells. Arterioscler Thromb Vasc Biol 2009; 29:1119-24. [PMID: 19372461 PMCID: PMC2704050 DOI: 10.1161/atvbaha.109.186239] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Vascular endothelial growth factor receptor 2 (VEGFR2) is a receptor tyrosine kinase that regulates vascular physiology. However, mechanism(s) by which VEGFR2 signaling and trafficking is coordinated are not clear. Here, we have tested endocytic Rab GTPases for regulation of VEGFR2 trafficking and signaling linked to endothelial cell migration. METHODS AND RESULTS Quiescent VEGFR2 displays endosomal localization and colocalization with the Rab5a GTPase, an early endosome fusion regulator. Expression of GTP or GDP-bound Rab5a mutants block activated VEGFR2 trafficking and degradation. Manipulation of Rab7a GTPase activity associated with late endosomes using overexpression of wild-type or mutant proteins blocks activated VEGFR2 trafficking and degradation. Depletion of Rab7a decreased VEGFR2 Y1175 phosphorylation but increased p42/44 (pERK1/2) MAPK phosphorylation. Endothelial cell migration was increased by Rab5a depletion but decreased by Rab7a depletion. CONCLUSIONS Rab5a and Rab7a regulate VEGFR2 trafficking toward early and late endosomes. Our data suggest that VEGFR2-mediated regulation of endothelial function is dependent on different but specific Rab-mediated GTP hydrolysis activity required for endosomal trafficking.
Collapse
Affiliation(s)
- Helen M. Jopling
- Endothelial Cell Biology Unit, Institute for Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, UK
| | - Adam Odell
- Endothelial Cell Biology Unit, Institute for Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, UK
| | - Nigel M. Hooper
- Proteolysis Research Group, Institute for Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, UK
| | - Ian C. Zachary
- Centre for Cardiovascular Biology and Medicine, Rayne Institute, University College London, London WC1E 6JJ, UK
| | - John H. Walker
- Endothelial Cell Biology Unit, Institute for Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, UK
| | - Sreenivasan Ponnambalam
- Endothelial Cell Biology Unit, Institute for Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
37
|
Vanlandingham PA, Ceresa BP. Rab7 regulates late endocytic trafficking downstream of multivesicular body biogenesis and cargo sequestration. J Biol Chem 2009; 284:12110-24. [PMID: 19265192 PMCID: PMC2673280 DOI: 10.1074/jbc.m809277200] [Citation(s) in RCA: 327] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Revised: 03/04/2009] [Indexed: 11/06/2022] Open
Abstract
The small molecular weight G-protein RAB7 is localized to both early and late endosomes and has been shown to be critical for trafficking through the endocytic pathway. The role of RAB7 in the endocytic pathway has been controversial, with some groups reporting that it regulates trafficking from early to late endosomes and others ascribing its role to trafficking between late endosomes and lysosomes. In this study, we use RNA interference to identify the exact step RAB7 regulates in the movement of the epidermal growth factor receptor (EGFR) from the cell surface to the lysosome. In the absence of RAB7, trafficking of the EGF.EGFR complex through the early endosome to the late endosome/multivesicular body (LE/MVB) does not change, but exiting from the LE/MVB is blocked. Ultrastructural analysis reveals that RAB7 is not required for formation of intraluminal vesicles of the LE/MVB, since RAB7-deficient cells have an increased number of enlarged LE/MVBs densely packed with intraluminal vesicles. Biochemical data indicate that the EGFR complex is sequestered in these intraluminal vesicles. Together, these data provide evidence that RAB7 is required for the transfer of cargo from the LE/MVB to the lysosome and for endocytic organelle maintenance.
Collapse
Affiliation(s)
- Phillip A Vanlandingham
- Department of Cell Biology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73126, USA
| | | |
Collapse
|
38
|
Abstract
Clathrin-mediated endocytosis sorts for degradation of more than 50 different growth factor receptors capable of relaying growth and differentiation signals by means of their cytoplasm-facing, intrinsic tyrosine kinase activity. The kinetics and alternative routings of receptor endocytosis critically regulate growth factor signaling, which underscores the importance of understanding mechanisms underlying fail-safe operation (robustness) and fidelity of the pathway. Like other robust systems, a layered hub-centric network controls receptor endocytosis. Characteristically, the modular hubs (e.g., AP2-Eps15 and Hrs) contain a membrane-anchoring lipid-binding domain, an ubiquitin-binding module, which recruits ubiquitinylated cargo, and a machinery enabling homo-assembly. Scheduled hub transitions, as well as cascades of Rab family guanosine triphosphatases and membrane bending machineries, define points of commitment to vesicle budding, thereby securing unidirectional trafficking. System's bistability permits stimulation by a growth factor, which oscillates a series of switches based on posttranslational protein modifications (i.e., phosphorylation, ubiquitinylation and neddylation), as well as transient low-affinity/high-avidity protein assemblies. Cbl family ubiquitin ligases, along with a set of phosphotyrosine-binding adaptors (e.g., Grb2), integrate receptor endocytosis into the densely wired networks of signal transduction pathways, which are involved in health and disease.
Collapse
Affiliation(s)
- Yaara Zwang
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
39
|
Semerdjieva S, Shortt B, Maxwell E, Singh S, Fonarev P, Hansen J, Schiavo G, Grant BD, Smythe E. Coordinated regulation of AP2 uncoating from clathrin-coated vesicles by rab5 and hRME-6. ACTA ACUST UNITED AC 2008; 183:499-511. [PMID: 18981233 PMCID: PMC2575790 DOI: 10.1083/jcb.200806016] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Here we investigate the role of rab5 and its cognate exchange factors rabex-5 and hRME-6 in the regulation of AP2 uncoating from endocytic clathrin-coated vesicles (CCVs). In vitro, we show that the rate of AP2 uncoating from CCVs is dependent on the level of functional rab5. In vivo, overexpression of dominant-negative rab5(S34N), or small interfering RNA (siRNA)-mediated depletion of hRME-6, but not rabex-5, resulted in increased steady-state levels of AP2 associated with endocytic vesicles, which is consistent with reduced uncoating efficiency. hRME-6 guanine nucleotide exchange factor activity requires hRME-6 binding to alpha-adaptin ear, which displaces the ear-associated mu2 kinase AAK1. siRNA-mediated depletion of hRME-6 increases phospho-mu2 levels, and expression of a phosphomimetic mu2 mutant increases levels of endocytic vesicle-associated AP2. Depletion of hRME-6 or rab5(S35N) expression also increases the levels of phosphoinositide 4,5-bisphosphate (PtdIns(4,5)P(2)) associated with endocytic vesicles. These data are consistent with a model in which hRME-6 and rab5 regulate AP2 uncoating in vivo by coordinately regulating mu2 dephosphorylation and PtdIns(4,5)P(2) levels in CCVs.
Collapse
Affiliation(s)
- Sophia Semerdjieva
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, England, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Leonard D, Hayakawa A, Lawe D, Lambright D, Bellve KD, Standley C, Lifshitz LM, Fogarty KE, Corvera S. Sorting of EGF and transferrin at the plasma membrane and by cargo-specific signaling to EEA1-enriched endosomes. J Cell Sci 2008; 121:3445-58. [PMID: 18827013 PMCID: PMC2586290 DOI: 10.1242/jcs.031484] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The biological function of receptors is determined by their appropriate trafficking through the endosomal pathway. Following internalization, the transferrin (Tf) receptor quantitatively recycles to the plasma membrane, whereas the epidermal growth factor (EGF) receptor undergoes degradation. To determine how Tf and EGF engage these two different pathways we imaged their binding and early endocytic pathway in live cells using total internal reflection fluorescence microscopy (TIRF-M). We find that EGF and Tf bind to distinct plasma membrane regions and are incorporated into different endocytic vesicles. After internalization, both EGF-enriched and Tf-enriched vesicles interact with endosomes containing early endosome antigen 1 (EEA1). EGF is incorporated and retained in these endosomes, while Tf-containing vesicles rapidly dissociate and move to a juxtanuclear compartment. Endocytic vesicles carrying EGF recruit more Rab5 GTPase than those carrying Tf, which, by strengthening their association with EEA1-enriched endosomes, may provide a mechanism for the observed cargo-specific sorting. These results reveal pre-endocytic sorting of Tf and EGF, a specialized role for EEA1-enriched endosomes in EGF trafficking, and a potential mechanism for cargo-specified sorting of endocytic vesicles by these endosomes.
Collapse
Affiliation(s)
- Deborah Leonard
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Hyatt DC, Ceresa BP. Cellular localization of the activated EGFR determines its effect on cell growth in MDA-MB-468 cells. Exp Cell Res 2008; 314:3415-25. [PMID: 18817771 DOI: 10.1016/j.yexcr.2008.08.020] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Revised: 08/25/2008] [Accepted: 08/26/2008] [Indexed: 01/23/2023]
Abstract
The epidermal growth factor (EGF) receptor (EGFR) is a ubiquitously expressed receptor tyrosine kinase that regulates diverse cell functions that are dependent upon cell type, the presence of downstream effectors, and receptor density. In addition to activating biochemical pathways, ligand stimulation causes the EGFR to enter the cell via clathrin-coated pits. Endocytic trafficking influences receptor signaling by controlling the duration of EGFR phosphorylation and coordinating the receptor's association with downstream effectors. To better understand the individual contributions of cell surface and cytosolic EGFRs on cell physiology, we used EGF that was conjugated to 900 nm polystyrene beads (EGF-beads). EGF-beads can stimulate the EGFR and retain the activated receptor at the plasma membrane. In MDA-MB-468 cells, a breast cancer cell line that over-expresses the EGFR, only internalized, activated EGFRs stimulate caspase-3 and induce cell death. Conversely, signaling cascades triggered from activated EGFR retained at the cell surface inhibit caspase-3 and promote cell proliferation. Thus, through endocytosis, the activated EGFR can differentially regulate cell growth in MDA-MB-468 cells.
Collapse
Affiliation(s)
- Dustin C Hyatt
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd, Oklahoma City, OK 73190, USA
| | | |
Collapse
|
42
|
Deng Q, Barbieri JT. Modulation of host cell endocytosis by the type III cytotoxin, Pseudomonas ExoS. Traffic 2008; 9:1948-57. [PMID: 18778330 DOI: 10.1111/j.1600-0854.2008.00808.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Pseudomonas aeruginosa ExoS is a bifunctional type III cytotoxin that possesses Rho GTPase-activating protein (RhoGAP) and ADP-ribosyltransferase (ADPr) activities. In the current study, the RhoGAP and ADPr activities of ExoS were tested for the ability to disrupt mammalian epithelial cell physiology. RhoGAP, but not ADPr, inhibited internalization/phagocytosis of bacteria, while ADPr, but not RhoGAP, inhibited vesicle trafficking, both general fluid-phase uptake and EGF-activated EGF receptor (EGFR) degradation. In ADPr-intoxicated cells, upon EGF activation, EGFR co-localized with clathrin-coated vesicles (CCV), which did not mature into Rab5-positive early endosomes. Constitutively, active Rab5 recruited EGFR from CCV to early endosomes. Consistent with the inhibition of Rab5 function by ADPr, several Rab proteins including Rab5 and 9, but not Rab4, were ADP ribosylated by ExoS. Thus, the two enzymatic activities of ExoS have different effects on epithelial cells with RhoGAP inhibiting bacterial internalization and ADPr interfering with CCV maturation. The ability ADPr to inhibit mammalian vesicle trafficking provides a new mechanism for bacterial toxin-mediated virulence.
Collapse
Affiliation(s)
- Qing Deng
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | | |
Collapse
|
43
|
Dhani SU, Kim Chiaw P, Huan LJ, Bear CE. ATP depletion inhibits the endocytosis of ClC-2. J Cell Physiol 2007; 214:273-80. [PMID: 17620322 DOI: 10.1002/jcp.21192] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The chloride channel, ClC-2 is expressed ubiquitously and participates in multiple physiological processes. In particular, ClC-2 has been implicated in the regulation of neuronal chloride ion homeostasis and mutations in ClC-2 are associated with idiopathic generalized epilepsy. Despite the physiological and pathophysiological significance of this channel, its regulation remains incompletely understood. The functional expression of ClC-2 at the cell surface has been shown to be enhanced by depletion of cellular ATP, implicating its possible role in cellular energy sensing. In the present study, biochemical assays of cell surface expression suggest that this gain of function reflects, in part, an increase in channel number due to the reduction in ClC-2 internalization by endocytosis. Cell surface expression of the disease-causing mutant: G715E, thought to lack wild-type nucleotide binding affinity, is similarly affected, suggesting that ATP-depletion modifies the function of proteins in the endocytic pathway rather than ClC-2 directly. Using a combination of immunofluorescence and biochemical studies, we confirmed that ClC-2 is internalized via dynamin-dependent endocytosis and that the change in surface expression evoked by ATP depletion is partially mimicked by inhibition of dynamin function using a dynamin dominant-negative mutant (DynK44A). Furthermore, trafficking via the early endosomal compartment occurs in part through rab5-associated vesicles and recycling of ClC-2 to the cell surface occurs through a rab11 dependent pathway. In summary, we have determined that the internalization of ClC-2 by endocytosis is inhibited by metabolic stress, highlighting the importance for understanding the molecular mechanisms mediating the endosomal trafficking of this channel.
Collapse
Affiliation(s)
- Sonja U Dhani
- Programme in Molecular Structure and Function, Research Institute in the Hospital for Sick Children, Toronto, Canada
| | | | | | | |
Collapse
|
44
|
Wong AW, Scales SJ, Reilly DE. DNA Internalized via Caveolae Requires Microtubule-dependent, Rab7-independent Transport to the Late Endocytic Pathway for Delivery to the Nucleus. J Biol Chem 2007; 282:22953-63. [PMID: 17562704 DOI: 10.1074/jbc.m611015200] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Using cationic liposomes to mediate gene delivery by transfection has the advantages of improved safety and simplicity of use over viral gene therapy. Understanding the mechanism by which cationic liposome:DNA complexes are internalized and delivered to the nucleus should help identify which transport steps might be manipulated in order to improve transfection efficiencies. We therefore examined the endocytosis and trafficking of two cationic liposomes, DMRIE-C and Lipofectamine LTX, in CHO cells. We found that DMRIE-C-transfected DNA is internalized via caveolae, while LTX-transfected DNA is internalized by clathrin-mediated endocytosis, with both pathways converging at the late endosome or lysosome. Inhibition of microtubule-dependent transport with nocodazole revealed that DMRIE-C:DNA complexes cannot enter the cytosol directly from caveosomes. Lysosomal degradation of transfected DNA has been proposed to be a major reason for poor transfection efficiency. However, in our system dominant negatives of both Rab7 and its effector RILP inhibited late endosome to lysosome transport of DNA complexes and LDL, but did not affect DNA delivery to the nucleus. This suggests that DNA is able to escape from late endosomes without traversing lysosomes and that caveosome to late endosome transport does not require Rab7 function. Lysosomal inhibition with chloroquine likewise had no effect on transfection product titers. These data suggest that DMRIE-C and LTX transfection complexes are endocytosed by separate pathways that converge at the late endosome or lysosome, but that blocking lysosomal traffic does not improve transfection product yields, identifying late endosome/lysosome to nuclear delivery as a step for future study.
Collapse
Affiliation(s)
- Athena W Wong
- Department of Early Stage Cell Culture, Genentech Inc., South San Francisco, California 94080, USA.
| | | | | |
Collapse
|
45
|
Fu D, van Dam EM, Brymora A, Duggin IG, Robinson PJ, Roufogalis BD. The small GTPases Rab5 and RalA regulate intracellular traffic of P-glycoprotein. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:1062-72. [PMID: 17524504 DOI: 10.1016/j.bbamcr.2007.03.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2006] [Revised: 02/28/2007] [Accepted: 03/27/2007] [Indexed: 12/11/2022]
Abstract
P-glycoprotein (P-gp) is a plasma membrane glycoprotein that can cause multidrug resistance (MDR) of cancer cells by acting as an ATP-dependent drug efflux pump. The regulatory effects of the small GTPases Rab5 and RalA on the intracellular trafficking of P-gp were investigated in HeLa cells. As expected, overexpressed enhanced green fluorescent protein (EGFP)-tagged P-gp (P-gp-EGFP) is mainly localised to the plasma membrane. However, upon cotransfection of either dominant negative Rab5 (Rab5-S34N) or constitutively active RalA (RalA-G23V) the intracellular P-gp-EGFP levels increased approximately 9 and 13 fold, respectively, compared to control P-gp-EGFP cells. These results suggest that Rab5 and RalA regulate P-gp trafficking between the plasma membrane and an intracellular compartment. In contrast, coexpression of constitutively active Rab5 (Rab5-Q79L) or dominant negative RalA (RalA-S28N) had no effect on the localisation of P-gp-EGFP. Furthermore, the intracellular accumulation of daunorubicin, a substrate for P-gp, increased significantly with an increased intracellular localisation of P-gp-EGFP. These results imply that it may be possible to overcome MDR by controlling the plasma membrane localisation of P-gp.
Collapse
Affiliation(s)
- Dong Fu
- Faculty of Pharmacy, The University of Sydney, NSW 2006, Australia
| | | | | | | | | | | |
Collapse
|
46
|
Zhang J, Schulze KL, Hiesinger PR, Suyama K, Wang S, Fish M, Acar M, Hoskins RA, Bellen HJ, Scott MP. Thirty-one flavors of Drosophila rab proteins. Genetics 2007; 176:1307-22. [PMID: 17409086 PMCID: PMC1894592 DOI: 10.1534/genetics.106.066761] [Citation(s) in RCA: 247] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Rab proteins are small GTPases that play important roles in transport of vesicle cargo and recruitment, association of motor and other proteins with vesicles, and docking and fusion of vesicles at defined locations. In vertebrates, >75 Rab genes have been identified, some of which have been intensively studied for their roles in endosome and synaptic vesicle trafficking. Recent studies of the functions of certain Rab proteins have revealed specific roles in mediating developmental signal transduction. We have begun a systematic genetic study of the 33 Rab genes in Drosophila. Most of the fly proteins are clearly related to specific vertebrate proteins. We report here the creation of a set of transgenic fly lines that allow spatially and temporally regulated expression of Drosophila Rab proteins. We generated fluorescent protein-tagged wild-type, dominant-negative, and constitutively active forms of 31 Drosophila Rab proteins. We describe Drosophila Rab expression patterns during embryogenesis, the subcellular localization of some Rab proteins, and comparisons of the localization of wild-type, dominant-negative, and constitutively active forms of selected Rab proteins. The high evolutionary conservation and low redundancy of Drosophila Rab proteins make these transgenic lines a useful tool kit for investigating Rab functions in vivo.
Collapse
Affiliation(s)
- Jun Zhang
- Departments of Developmental Biology, Genetics, and Bioengineering, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California 94305, Howard Hughes Medical Institute, Department of Molecular and Human Genetics, Stanford University School of Medicine, Stanford, California 94305, Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, Department of Genome Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720-3200 and Department of Physiology Green Center Division for Systems Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040
| | - Karen L. Schulze
- Departments of Developmental Biology, Genetics, and Bioengineering, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California 94305, Howard Hughes Medical Institute, Department of Molecular and Human Genetics, Stanford University School of Medicine, Stanford, California 94305, Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, Department of Genome Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720-3200 and Department of Physiology Green Center Division for Systems Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040
| | - P. Robin Hiesinger
- Departments of Developmental Biology, Genetics, and Bioengineering, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California 94305, Howard Hughes Medical Institute, Department of Molecular and Human Genetics, Stanford University School of Medicine, Stanford, California 94305, Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, Department of Genome Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720-3200 and Department of Physiology Green Center Division for Systems Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040
| | - Kaye Suyama
- Departments of Developmental Biology, Genetics, and Bioengineering, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California 94305, Howard Hughes Medical Institute, Department of Molecular and Human Genetics, Stanford University School of Medicine, Stanford, California 94305, Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, Department of Genome Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720-3200 and Department of Physiology Green Center Division for Systems Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040
| | - Stream Wang
- Departments of Developmental Biology, Genetics, and Bioengineering, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California 94305, Howard Hughes Medical Institute, Department of Molecular and Human Genetics, Stanford University School of Medicine, Stanford, California 94305, Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, Department of Genome Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720-3200 and Department of Physiology Green Center Division for Systems Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040
| | - Matthew Fish
- Departments of Developmental Biology, Genetics, and Bioengineering, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California 94305, Howard Hughes Medical Institute, Department of Molecular and Human Genetics, Stanford University School of Medicine, Stanford, California 94305, Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, Department of Genome Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720-3200 and Department of Physiology Green Center Division for Systems Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040
| | - Melih Acar
- Departments of Developmental Biology, Genetics, and Bioengineering, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California 94305, Howard Hughes Medical Institute, Department of Molecular and Human Genetics, Stanford University School of Medicine, Stanford, California 94305, Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, Department of Genome Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720-3200 and Department of Physiology Green Center Division for Systems Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040
| | - Roger A. Hoskins
- Departments of Developmental Biology, Genetics, and Bioengineering, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California 94305, Howard Hughes Medical Institute, Department of Molecular and Human Genetics, Stanford University School of Medicine, Stanford, California 94305, Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, Department of Genome Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720-3200 and Department of Physiology Green Center Division for Systems Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040
| | - Hugo J. Bellen
- Departments of Developmental Biology, Genetics, and Bioengineering, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California 94305, Howard Hughes Medical Institute, Department of Molecular and Human Genetics, Stanford University School of Medicine, Stanford, California 94305, Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, Department of Genome Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720-3200 and Department of Physiology Green Center Division for Systems Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040
| | - Matthew P. Scott
- Departments of Developmental Biology, Genetics, and Bioengineering, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California 94305, Howard Hughes Medical Institute, Department of Molecular and Human Genetics, Stanford University School of Medicine, Stanford, California 94305, Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, Department of Genome Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720-3200 and Department of Physiology Green Center Division for Systems Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040
- Corresponding author: Departments of Developmental Biology, Genetics, and Bioengineering, Howard Hughes Medical Institute, Clark Center, West Wing W252, 318 Campus Dr., Stanford University School of Medicine, Stanford, CA 94305-5439. E-mail:
| |
Collapse
|
47
|
Sossa KG, Court BL, Carroll RC. NMDA receptors mediate calcium-dependent, bidirectional changes in dendritic PICK1 clustering. Mol Cell Neurosci 2006; 31:574-85. [PMID: 16406232 DOI: 10.1016/j.mcn.2005.11.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2005] [Revised: 11/07/2005] [Accepted: 11/22/2005] [Indexed: 02/03/2023] Open
Abstract
AMPA receptor (AMPAR) trafficking at CNS synapses is regulated by several receptor-binding proteins. One model of AMPAR endocytosis entails the cotargeting of the GluR2-interacting protein PICK1 and activated PKC to synapses. We demonstrate that NMDA receptor (NMDAR) activation mediates bidirectional changes in surface AMPARs through two additional forms of PICK1 redistribution. In neurons, NMDAR activation, which induces AMPAR endocytosis, increases endosomal PICK1 clustering. In contrast, stronger NMDAR activation rapidly reduces PICK1 clustering accompanied by decreases in PICK1/GluR2 association and increases in surface AMPAR levels. PICK1-siRNA similarly increases surface AMPARs and occludes the NMDAR-mediated effect, demonstrating the role of PICK1 in this process. Bidirectional NMDAR-mediated changes in PICK1 localization are determined by the magnitude of receptor-activated dendritic calcium signals. Our results show that PICK1 localization in dendrites is subject to multiple forms of regulation that contribute to surface AMPAR expression, likely by modulating the numbers of AMPARs maintained in intracellular compartments.
Collapse
Affiliation(s)
- K G Sossa
- Department of Neuroscience, Rose Kennedy Center for Mental Retardation, 1410 Pelham Parkway, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | |
Collapse
|
48
|
Ceresa BP, Bahr SJ. rab7 Activity Affects Epidermal Growth Factor:Epidermal Growth Factor Receptor Degradation by Regulating Endocytic Trafficking from the Late Endosome. J Biol Chem 2006; 281:1099-106. [PMID: 16282324 DOI: 10.1074/jbc.m504175200] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is a member of the receptor tyrosine kinase family. Ligand (epidermal growth factor or EGF) binding to the EGFR results in the coordinated activation and integration of biochemical signaling events to mediate cell growth, migration, and differentiation. One mechanism the cell utilizes to orchestrate these events is ligand-mediated endocytosis through the canonical clathrin-mediated endocytic pathway. Identification of proteins that regulate the intracellular movement of the EGF.EGFR complex is an important first step in dissecting how specificity of EGFR signaling is conferred. We examined the role of the small molecular weight guanine nucleotide-binding protein (G-protein) rab7 as a regulator of the distal stages of the endocytic pathway. Through the transient expression of activating and inactivating mutants of rab7 in HeLa cells, we have determined that rab7 activity directly correlates with the rate of radiolabeled EGF and EGFR degradation. Furthermore, when inhibitory mutants of rab7 are expressed, the internalized EGF.EGFR complex accumulates in high-density endosomes that are characteristic of the late endocytic pathway. Thus, we conclude that rab7 regulates the endocytic trafficking of the EGF.EGFR complex by regulating its lysosomal degradation.
Collapse
Affiliation(s)
- Brian P Ceresa
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, 73190, USA.
| | | |
Collapse
|
49
|
Araripe JR, Ramos FP, Cunha e Silva NL, Urményi TP, Silva R, Leite Fontes CF, da Silveira JF, Rondinelli E. Characterization of a RAB5 homologue in Trypanosoma cruzi. Biochem Biophys Res Commun 2005; 329:638-45. [PMID: 15737633 DOI: 10.1016/j.bbrc.2005.02.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2005] [Indexed: 11/30/2022]
Abstract
RAB proteins are small GTPases involved in exocytic and endocytic pathways of eukaryotic cells, controlling vesicle docking and fusion. RABs show a remarkable specificity in subcellular localization, so they can be used as molecular markers for studying protein trafficking in Trypanosoma cruzi, the causal agent of Chagas' disease. RAB5 is a component of early endosomes. It has been identified in kinetoplastids such as Trypanosoma brucei and Leishmania donovani. In this work, we describe the characterization of the complete coding sequence of a RAB5 gene homologue in T. cruzi (TcRAB5, GenBank Accession No. AY730667). It is present as a single copy gene, located at chromosomal bands XIII and XIV. TcRAB5 shares the highest degrees of similarity (71%) and identity (63%) with Trypanosoma brucei rhodesiense RAB5a and contains all five characteristic RAB motifs. TcRAB5 is transcribed as a single 1.5kb mRNA in epimastigotes. Its transcript was also detected in the other two forms of the parasite, metacyclic trypomastigotes and spheromastigotes. The recombinant TcRAB5 protein was able to bind and hydrolyze GTP. The identification of proteins involved in T. cruzi endo- and exocytic pathways may generate cellular compartment markers, an invaluable tool to better understand the vesicular transport in this parasite.
Collapse
Affiliation(s)
- Júlia Rolão Araripe
- Laboratório de Metabolismo Macromolecular Firmino Torres de Castro, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, CCS, Cidade Universitária, Rio de Janeiro 21949-900, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Simpson JC, Griffiths G, Wessling-Resnick M, Fransen JAM, Bennett H, Jones AT. A role for the small GTPase Rab21 in the early endocytic pathway. J Cell Sci 2004; 117:6297-311. [PMID: 15561770 DOI: 10.1242/jcs.01560] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Rab proteins comprise a family of monomeric GTPases that control cellular membrane traffic. Rab21 is a poorly characterised member with no known function. Human Rab21 cDNA from K562 cells was subcloned into GFP expression vectors to generate Rab21 and Rab21 mutants defective in either GTP hydrolysis (Rab21 Q78L) or binding (Rab21 T33N) for transfection studies in HeLa cells. Confocal fluorescence microscopy and ultrastructural studies revealed Rab21 to be predominantly localised to the early endocytic pathway, on vesicles containing earlyendosomal antigen 1 EEA1, transferrin receptor and internalised ligands. EEA1 was localised to enlarged endosomes in Rab21 wild-type expressing cells but the GTP hydrolysis and GDP binding mutants had unique phenotypes labelling tubular reticular structures and the trans-Golgi network, respectively. Early endosome localisation for Rab21 was confirmed in a hepatoma cell line that allowed analysis of the subcellular distribution of the endogenous protein. Comparison of the localisation of Rab21 with other Rabs revealed extensive colocalisation with early endocytic variants Rab4, Rab5, Rab17 and Rab22 but much less overlap with those associated with late endosomes, recycling endosomes and the early secretory pathway. Cells expressing Rab21 T33N had defects in endocytosis of transferrin and epidermal growth factor and failed to effectively deliver the latter ligand to late endosomes and lysosomes for degradation. Collectively, our data provide the first characterisation of Rab21 function in early endosome dynamics.
Collapse
Affiliation(s)
- Jeremy C Simpson
- Cell Biology and Biophysics Programme, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|