1
|
Bowman DM, Meenderink LM, Thomas KS, Manning EH, Tyska MJ, Goldenring JR. Microvillus inclusion disease-causing MYO5B point mutations exert differential effects on motor function. J Biol Chem 2025; 301:108328. [PMID: 39978676 PMCID: PMC11964754 DOI: 10.1016/j.jbc.2025.108328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/12/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
Microvillus inclusion disease (MVID) is a rare congenital diarrheal disorder typically caused by loss of function mutations in the unconventional myosin, myosin 5b (MYO5B), which leads to the mistrafficking of apical components in enterocytes. MVID can manifest in two phenotypes: in both the intestine and liver or the liver alone. Although previous studies seeking to understand MVID disease pathology used MYO5B KO models, many patients have point mutations and thus express a dysfunctional MYO5B. How these point mutations lead to a broad spectrum of disease severity and the development of two distinct disease phenotypes is still not known. Here, we investigate the effect of MVID patient mutations on the function of the MYO5B motor domain, independent of cargo binding, using confocal imaging and fluorescence recovery after photobleaching. Patient mutations demonstrated a range of effects in these assays, from rigor-like behavior to loss of actin binding. Additionally, analysis of fluorescence recovery after photobleaching turnover kinetics suggests that some mutations negatively impact the ability of MYO5B to stay bound to actin. Collectively, our findings indicate that patient mutations affect the MYO5B motor domain in diverse ways, consistent with the spectrum of phenotypes observed in patients.
Collapse
Affiliation(s)
- Deanna M Bowman
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Leslie M Meenderink
- Division of Infectious Disease, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Veterans Affairs Tennessee Valley Health Care System, Nashville, Tennessee, USA
| | - Kyra S Thomas
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Section of Surgical Science, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Elizabeth H Manning
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Veterans Affairs Tennessee Valley Health Care System, Nashville, Tennessee, USA; Section of Surgical Science, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James R Goldenring
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Veterans Affairs Tennessee Valley Health Care System, Nashville, Tennessee, USA; Section of Surgical Science, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| |
Collapse
|
2
|
Kisler K, Sagare AP, Lazic D, Bazzi S, Lawson E, Hsu CJ, Wang Y, Ramanathan A, Nelson AR, Zhao Z, Zlokovic BV. Anti-malaria drug artesunate prevents development of amyloid-β pathology in mice by upregulating PICALM at the blood-brain barrier. Mol Neurodegener 2023; 18:7. [PMID: 36707892 PMCID: PMC9883925 DOI: 10.1186/s13024-023-00597-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/13/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND PICALM is one of the most significant susceptibility factors for Alzheimer's disease (AD). In humans and mice, PICALM is highly expressed in brain endothelium. PICALM endothelial levels are reduced in AD brains. PICALM controls several steps in Aβ transcytosis across the blood-brain barrier (BBB). Its loss from brain endothelium in mice diminishes Aβ clearance at the BBB, which worsens Aβ pathology, but is reversible by endothelial PICALM re-expression. Thus, increasing PICALM at the BBB holds potential to slow down development of Aβ pathology. METHODS To identify a drug that could increase PICALM expression, we screened a library of 2007 FDA-approved drugs in HEK293t cells expressing luciferase driven by a human PICALM promoter, followed by a secondary mRNA screen in human Eahy926 endothelial cell line. In vivo studies with the lead hit were carried out in Picalm-deficient (Picalm+/-) mice, Picalm+/-; 5XFAD mice and Picalmlox/lox; Cdh5-Cre; 5XFAD mice with endothelial-specific Picalm knockout. We studied PICALM expression at the BBB, Aβ pathology and clearance from brain to blood, cerebral blood flow (CBF) responses, BBB integrity and behavior. RESULTS Our screen identified anti-malaria drug artesunate as the lead hit. Artesunate elevated PICALM mRNA and protein levels in Eahy926 endothelial cells and in vivo in brain capillaries of Picalm+/- mice by 2-3-fold. Artesunate treatment (32 mg/kg/day for 2 months) of 3-month old Picalm+/-; 5XFAD mice compared to vehicle increased brain capillary PICALM levels by 2-fold, and reduced Aβ42 and Aβ40 levels and Aβ and thioflavin S-load in the cortex and hippocampus, and vascular Aβ load by 34-51%. Artesunate also increased circulating Aβ42 and Aβ40 levels by 2-fold confirming accelerated Aβ clearance from brain to blood. Consistent with reduced Aβ pathology, treatment of Picalm+/-; 5XFAD mice with artesunate improved CBF responses, BBB integrity and behavior on novel object location and recognition, burrowing and nesting. Endothelial-specific knockout of PICALM abolished all beneficial effects of artesunate in 5XFAD mice indicating that endothelial PICALM is required for its therapeutic effects. CONCLUSIONS Artesunate increases PICALM levels and Aβ clearance at the BBB which prevents development of Aβ pathology and functional deficits in mice and holds potential for translation to human AD.
Collapse
Affiliation(s)
- Kassandra Kisler
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, 1501 San Pablo St, Los Angeles, CA 90089 USA
| | - Abhay P. Sagare
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, 1501 San Pablo St, Los Angeles, CA 90089 USA
| | - Divna Lazic
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, 1501 San Pablo St, Los Angeles, CA 90089 USA
| | - Sam Bazzi
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, 1501 San Pablo St, Los Angeles, CA 90089 USA
| | - Erica Lawson
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, 1501 San Pablo St, Los Angeles, CA 90089 USA
| | - Ching-Ju Hsu
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, 1501 San Pablo St, Los Angeles, CA 90089 USA
| | - Yaoming Wang
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, 1501 San Pablo St, Los Angeles, CA 90089 USA
| | - Anita Ramanathan
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, 1501 San Pablo St, Los Angeles, CA 90089 USA
| | - Amy R. Nelson
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, 1501 San Pablo St, Los Angeles, CA 90089 USA
| | - Zhen Zhao
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, 1501 San Pablo St, Los Angeles, CA 90089 USA
| | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, 1501 San Pablo St, Los Angeles, CA 90089 USA
| |
Collapse
|
3
|
Abstract
Vesicles mediate the trafficking of membranes/proteins in the endocytic and secretory pathways. These pathways are regulated by small GTPases of the Rab family. Rab proteins belong to the Ras superfamily of GTPases, which are significantly involved in various intracellular trafficking and signaling processes in the nervous system. Rab11 is known to play a key role especially in recycling many proteins, including receptors important for signal transduction and preservation of functional activities of nerve cells. Rab11 activity is controlled by GEFs (guanine exchange factors) and GAPs (GTPase activating proteins), which regulate its function through modulating GTP/GDP exchange and the intrinsic GTPase activity, respectively. Rab11 is involved in the transport of several growth factor molecules important for the development and repair of neurons. Overexpression of Rab11 has been shown to significantly enhance vesicle trafficking. On the other hand, a reduced expression of Rab11 was observed in several neurodegenerative diseases. Current evidence appears to support the notion that Rab11 and its cognate proteins may be potential targets for therapeutic intervention. In this review, we briefly discuss the function of Rab11 and its related interaction partners in intracellular pathways that may be involved in neurodegenerative processes.
Collapse
Affiliation(s)
| | - Jiri Novotny
- Jiri Novotny, Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic.
| |
Collapse
|
4
|
Rathan-Kumar S, Roland JT, Momoh M, Goldstein A, Lapierre LA, Manning E, Mitchell L, Norman J, Kaji I, Goldenring JR. Rab11FIP1-deficient mice develop spontaneous inflammation and show increased susceptibility to colon damage. Am J Physiol Gastrointest Liver Physiol 2022; 323:G239-G254. [PMID: 35819177 PMCID: PMC9423785 DOI: 10.1152/ajpgi.00042.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/23/2022] [Accepted: 06/29/2022] [Indexed: 01/31/2023]
Abstract
The small GTPase, Rab11a, regulates vesicle trafficking and cell polarity in epithelial cells through interaction with Rab11 family-interacting proteins (Rab11-FIPs). We hypothesized that deficiency of Rab11-FIP1 would affect mucosal integrity in the intestine. Global Rab11FIP1 knockout (KO) mice were generated by deletion of the second exon. Pathology of intestinal tissues was analyzed by immunostaining of colonic sections and RNA-sequencing of isolated colonic epithelial cells. A low concentration of dextran sodium sulfate (DSS, 2%) was added to drinking water for 5 days, and injury score was compared between Rab11FIP1 KO, Rab11FIP2 KO, and heterozygous littermates. Rab11FIP1 KO mice showed normal fertility and body weight gain. More frequent lymphoid patches and infiltration of macrophages and neutrophils were identified in Rab11FIP1 KO mice before the development of rectal prolapse compared with control mice. The population of trefoil factor 3 (TFF3)-positive goblet cells was significantly lower, and the ratio of proliferative to nonproliferative cells was higher in Rab11FIP1 KO colons. Transcription signatures indicated that Rab11FIP1 deletion downregulated genes that mediate stress tolerance response, whereas genes mediating the response to infection were significantly upregulated, consistent with the inflammatory responses in the steady state. Lack of Rab11FIP1 also resulted in abnormal accumulation of subapical vesicles in colonocytes and the internalization of transmembrane mucin, MUC13, with Rab14. After DSS treatment, Rab11FIP1 KO mice showed greater body weight loss and more severe mucosal damage than those in heterozygous littermates. These findings suggest that Rab11FIP1 is important for cytoprotection mechanisms and for the maintenance of colonic mucosal integrity.NEW & NOTEWORTHY Although Rab11FIP1 is important in membrane trafficking in epithelial cells, the gastrointestinal phenotype of Rab11FIP1 knockout (KO) mice had never been reported. This study demonstrated that Rab11FIP1 loss induces mistrafficking of Rab14 and MUC13 and decreases in colonic goblet cells, resulting in impaired mucosal integrity.
Collapse
Affiliation(s)
- Sudiksha Rathan-Kumar
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joseph T Roland
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Michael Momoh
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anna Goldstein
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lynne A Lapierre
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Elizabeth Manning
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Louise Mitchell
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
| | - Jim Norman
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
| | - Izumi Kaji
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - James R Goldenring
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee
| |
Collapse
|
5
|
Goswami S, Balasubramanian I, D'Agostino L, Bandyopadhyay S, Patel R, Avasthi S, Yu S, Goldenring JR, Bonder EM, Gao N. RAB11A-mediated YAP localization to adherens and tight junctions is essential for colonic epithelial integrity. J Biol Chem 2021; 297:100848. [PMID: 34058200 PMCID: PMC8254046 DOI: 10.1016/j.jbc.2021.100848] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 05/21/2021] [Accepted: 05/27/2021] [Indexed: 12/16/2022] Open
Abstract
Within the intestinal epithelium, regulation of intracellular protein and vesicular trafficking is of utmost importance for barrier maintenance, immune responses, and tissue polarity. RAB11A is a small GTPase that mediates the anterograde transport of protein cargos to the plasma membrane. Loss of RAB11A-dependent trafficking in mature intestinal epithelial cells results in increased epithelial proliferation and nuclear accumulation of Yes-associated protein (YAP), a key Hippo-signaling transducer that senses cell–cell contacts and regulates tissue growth. However, it is unclear how RAB11A regulates YAP intracellular localizations. In this report, we examined the relationship of RAB11A to epithelial junctional complexes, YAP, and the associated consequences on colonic epithelial tissue repair. We found that RAB11A controls the biochemical associations of YAP with multiple components of adherens and tight junctions, including α-catenin, β-catenin, and Merlin, a tumor suppressor. In the absence of RAB11A and Merlin, we observed enhanced YAP–β-catenin complex formation and nuclear translocation. Upon chemical injury to the intestine, mice deficient in RAB11A were found to have reduced epithelial integrity, decreased YAP localization to adherens and tight junctions, and increased nuclear YAP accumulation in the colon epithelium. Thus, RAB11A-regulated trafficking regulates the Hippo–YAP signaling pathway for rapid reparative response after tissue injury.
Collapse
Affiliation(s)
- Sayantani Goswami
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | | | - Luca D'Agostino
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | | | - Radha Patel
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Shail Avasthi
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Shiyan Yu
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - James R Goldenring
- Department of Surgery, Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Edward M Bonder
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA.
| |
Collapse
|
6
|
Datta A, Yang CR, Limbutara K, Chou CL, Rinschen MM, Raghuram V, Knepper MA. PKA-independent vasopressin signaling in renal collecting duct. FASEB J 2020; 34:6129-6146. [PMID: 32219907 PMCID: PMC9200475 DOI: 10.1096/fj.201902982r] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 02/13/2020] [Accepted: 02/19/2020] [Indexed: 11/11/2022]
Abstract
Vasopressin regulates renal water excretion by binding to a Gα s-coupled receptor (V2R) in collecting duct cells, resulting in increased water permeability through regulation of the aquaporin-2 (AQP2) water channel. This action is widely accepted to be associated with cAMP-mediated activation of protein kinase A (PKA). Here, we use phosphoproteomics in collecting duct cells in which PKA has been deleted (CRISPR-Cas9) to identify PKA-independent responses to vasopressin. The results show that V2R-mediated vasopressin signaling is predominantly, but not entirely, PKA-dependent. Upregulated sites in PKA-null cells include Ser256 of AQP2, which is critical to regulation of AQP2 trafficking. In addition, phosphorylation changes in the protein kinases Stk39 (SPAK) and Prkci (an atypical PKC) are consistent with PKA-independent regulation of these protein kinases. Target motif analysis of the phosphopeptides increased in PKA-null cells indicates that vasopressin activates one or more members of the AMPK/SNF1-subfamily of basophilic protein kinases. In vitro phosphorylation assays using recombinant, purified SNF1-subfamily kinases confirmed postulated target specificities. Of interest, measured IBMX-dependent cAMP levels were an order of magnitude higher in PKA-null than in PKA-intact cells, indicative of a PKA-dependent feedback mechanism. Overall, the findings support the conclusion that V2-receptor mediated signaling in collecting duct cells is in part PKA-independent.
Collapse
Affiliation(s)
- Arnab Datta
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
- Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore 575018, Karnataka, India
| | - Chin-Rang Yang
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Kavee Limbutara
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Chung-Lin Chou
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Markus M. Rinschen
- Department of Chemistry, Center for Metabolomics and Mass Spectrometry, The Scripps Research Institute, La Jolla, CA
| | - Viswanathan Raghuram
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Mark A. Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
7
|
Royo M, Gutiérrez Y, Fernández-Monreal M, Gutiérrez-Eisman S, Jiménez R, Jurado S, Esteban JA. A retention-release mechanism based on RAB11FIP2 for AMPA receptor synaptic delivery during long-term potentiation. J Cell Sci 2019; 132:jcs.234237. [PMID: 31757887 DOI: 10.1242/jcs.234237] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 11/15/2019] [Indexed: 02/05/2023] Open
Abstract
It is well--established that Rab11-dependent recycling endosomes drive the activity-dependent delivery of AMPA receptors (AMPARs) into synapses during long-term potentiation (LTP). Nevertheless, the molecular basis for this specialized function of recycling endosomes is still unknown. Here, we have investigated RAB11FIP2 (FIP2 hereafter) as a potential effector of Rab11-dependent trafficking during LTP in rat hippocampal slices. Surprisingly, we found that FIP2 operates independently from Rab11 proteins, and acts as a negative regulator of AMPAR synaptic trafficking. Under basal conditions, FIP2 associates with AMPARs at immobile compartments, separately from recycling endosomes. Using shRNA-mediated knockdown, we found that FIP2 prevents GluA1 (encoded by the Gria1 gene) AMPARs from reaching the surface of dendritic spines in the absence of neuronal stimulation. Upon induction of LTP, FIP2 is rapidly mobilized, dissociates from AMPARs and undergoes dephosphorylation. Interestingly, this dissociation of the FIP2-AMPAR complex, together with FIP2 dephosphorylation, is required for LTP, but the interaction between FIP2 and Rab11 proteins is not. Based on these results, we propose a retention-release mechanism, where FIP2 acts as a gate that restricts the trafficking of AMPARs, until LTP induction triggers their release and allows synaptic delivery.
Collapse
Affiliation(s)
- María Royo
- Department of Neurobiology, Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), 28049 Madrid, Spain
| | - Yolanda Gutiérrez
- Department of Neurobiology, Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), 28049 Madrid, Spain
| | - Mónica Fernández-Monreal
- Department of Neurobiology, Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), 28049 Madrid, Spain
| | - Silvia Gutiérrez-Eisman
- Department of Neurobiology, Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), 28049 Madrid, Spain
| | - Raquel Jiménez
- Department of Neurobiology, Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), 28049 Madrid, Spain
| | - Sandra Jurado
- Instituto de Neurociencias CSIC-UMH, 03550 San Juan de Alicante, Spain
| | - José A Esteban
- Department of Neurobiology, Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), 28049 Madrid, Spain
| |
Collapse
|
8
|
Dong W, Li H, Wu X. Rab11-FIP2 suppressed tumor growth via regulation of PGK1 ubiquitination in non-small cell lung cancer. Biochem Biophys Res Commun 2018; 508:60-65. [PMID: 30471866 DOI: 10.1016/j.bbrc.2018.11.108] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 11/16/2018] [Indexed: 02/07/2023]
Abstract
Mounting evidence has shown that the Rab11-FIP2 has critical roles in cancer cell growth. However, the clinical significance of Rab11-FIP2 in Non-small cell lung cancer (NSCLC) remains to be fully elucidated. In this study, we investigated the expression of Rab11-FIP2 using immunohistochemistry in 150 patients with NSCLC. We found that its expression level in NSCLC was much lower than that in the corresponding adjacent normal tissues. The DNA methylation data revealed that Rab11-FIP2 were significantly hypermethylated in NSCLC. The methylation level in the gene body was negatively correlated with the expression level of Rab11-FIP2 in NSCLC. Furthermore, enforced expression of Rab11-FIP2 dramatically reduced cancer cell proliferation and tumorigenesis, indicating a tumor suppressor role of PGK1 in NSCLC progression. Mechanistic investigations showed that Rab11-FIP2 interacted with the glycolytic kinase PGK1 and promoted its ubiquitination in NSCLC cells, leading to inactivation of the oncogenic AKT/mTOR signaling pathway. Overall, our data indicate that reduced expression of Rab11-FIP2 by DNA hypermethylation plays an important role in NSCLC tumor growth.
Collapse
Affiliation(s)
- Wenjie Dong
- Department of Internal Medicine-Oncology, The First Affiliated Hospital, Zhengzhou University, China.
| | - Huixia Li
- Department of Internal Medicine-Oncology, The First Affiliated Hospital, Zhengzhou University, China
| | - Xinai Wu
- Department of Internal Medicine-Oncology, The First Affiliated Hospital, Zhengzhou University, China.
| |
Collapse
|
9
|
Inhibition of the miR-192/215-Rab11-FIP2 axis suppresses human gastric cancer progression. Cell Death Dis 2018; 9:778. [PMID: 30006518 PMCID: PMC6045576 DOI: 10.1038/s41419-018-0785-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 06/07/2018] [Accepted: 06/11/2018] [Indexed: 12/17/2022]
Abstract
Less than a century ago, gastric cancer (GC) was the most common cancer throughout the world. Despite advances in surgical, chemotherapeutic, and radiotherapeutic treatment, GC remains the number 3 cancer killer worldwide. This fact highlights the need for better diagnostic biomarkers and more effective therapeutic targets. RAB11-FIP2, a member of the Rab11 family of interacting proteins, exhibits potential tumor suppressor function. However, involvement of RAB11-FIP2 in gastric carcinogenesis is yet to be elucidated. In this study, we demonstrated that RAB11-FIP2 was downregulated in GC tissues and constituted a target of the known onco-miRs, miR-192/215. We also showed that functionally, Rab11-FIP2 regulation by miR-192/215 is involved in GC-related biological activities. Finally, RAB11-FIP2 inhibition by miR-192/215 affected the establishment of cell polarity and tight junction formation in GC cells. In summary, this miR-192/215–Rab11-FIP2 axis appears to represent a new molecular mechanism underlying GC progression, while supplying a promising avenue of further research into diagnosis and therapy of GC.
Collapse
|
10
|
Jewett CE, Prekeris R. Insane in the apical membrane: Trafficking events mediating apicobasal epithelial polarity during tube morphogenesis. Traffic 2018; 19:10.1111/tra.12579. [PMID: 29766620 PMCID: PMC6239989 DOI: 10.1111/tra.12579] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 12/13/2022]
Abstract
The creation of cellular tubes is one of the most vital developmental processes, resulting in the formation of most organ types. Cells have co-opted a number of different mechanisms for tube morphogenesis that vary among tissues and organisms; however, generation and maintenance of cell polarity is fundamental for successful lumenogenesis. Polarized membrane transport has emerged as a key driver not only for establishing individual epithelial cell polarity, but also for coordination of epithelial polarization during apical lumen formation and tissue morphogenesis. In recent years, much work has been dedicated to identifying membrane trafficking regulators required for lumenogenesis. In this review we will summarize the findings from the past couple of decades in defining the molecular machinery governing lumenogenesis both in 3D tissue culture models and during organ development in vivo.
Collapse
Affiliation(s)
- Cayla E. Jewett
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rytis Prekeris
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
11
|
Warden CH, Bettaieb A, Min E, Fisler JS, Haj FG, Stern JS. Chow fed UC Davis strain female Lepr fatty Zucker rats exhibit mild glucose intolerance, hypertriglyceridemia, and increased urine volume, all reduced by a Brown Norway strain chromosome 1 congenic donor region. PLoS One 2017; 12:e0188175. [PMID: 29211750 PMCID: PMC5718614 DOI: 10.1371/journal.pone.0188175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 10/23/2017] [Indexed: 12/01/2022] Open
Abstract
Our objective is to identify genes that influence the development of any phenotypes of type 2 diabetes (T2D) or kidney disease in obese animals. We use the reproductively isolated UC Davis fatty Zucker strain rat model in which the defective chromosome 4 leptin receptor (LeprfaSte/faSte) results in fatty obesity. We previously produced a congenic strain with the distal half of chromosome 1 from the Brown Norway strain (BN) on a Zucker (ZUC) background (BN.ZUC-D1Rat183–D1Rat90). Previously published studies in males showed that the BN congenic donor region protects from some phenotypes of renal dysfunction and T2D. We now expand our studies to include females and expand phenotyping to gene expression. We performed diabetes and kidney disease phenotyping in chow-fed females of the BN.ZUC-D1Rat183-D1Rat90 congenic strain to determine the specific characteristics of the UC Davis model. Fatty LeprfaSte/faSte animals of both BN and ZUC genotype in the congenic donor region had prediabetic levels of fasting blood glucose and blood glucose 2 hours after a glucose tolerance test. We observed significant congenic strain chromosome 1 genotype effects of the BN donor region in fatty females that resulted in decreased food intake, urine volume, glucose area under the curve during glucose tolerance test, plasma triglyceride levels, and urine glucose excretion per day. In fatty females, there were significant congenic strain BN genotype effects on non-fasted plasma urea nitrogen, triglyceride, and creatinine. Congenic region genotype effects were observed by quantitative PCR of mRNA from the kidney for six genes, all located in the chromosome 1 BN donor region, with potential effects on T2D or kidney function. The results are consistent with the hypothesis that the BN genotype chromosome 1 congenic region influences traits of both type 2 diabetes and kidney function in fatty UC Davis ZUC females and that there are many positional candidate genes.
Collapse
Affiliation(s)
- Craig H. Warden
- Departments of Pediatrics, Neurobiology Physiology and Behavior, University of California, Davis, Davis, CA, United States of America
- * E-mail:
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee, Knoxville, TN, United States of America
| | - Esther Min
- Department of Nutrition, University of California, Davis, Davis, CA, United States of America
| | - Janis S. Fisler
- Department of Nutrition, University of California, Davis, Davis, CA, United States of America
| | - Fawaz G. Haj
- Department of Nutrition, University of California, Davis, Davis, CA, United States of America
| | - Judith S. Stern
- Department of Nutrition, University of California, Davis, Davis, CA, United States of America
- Internal Medicine, University of California, Davis, Davis, CA, United States of America
| |
Collapse
|
12
|
Garcia-Castillo MD, Chinnapen DJF, Lencer WI. Membrane Transport across Polarized Epithelia. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a027912. [PMID: 28213463 DOI: 10.1101/cshperspect.a027912] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Polarized epithelial cells line diverse surfaces throughout the body forming selective barriers between the external environment and the internal milieu. To cross these epithelial barriers, large solutes and other cargoes must undergo transcytosis, an endocytic pathway unique to polarized cell types, and significant for the development of cell polarity, uptake of viral and bacterial pathogens, transepithelial signaling, and immunoglobulin transport. Here, we review recent advances in our knowledge of the transcytotic pathway for proteins and lipids. We also discuss briefly the promise of harnessing the molecules that undergo transcytosis as vehicles for clinical applications in drug delivery.
Collapse
Affiliation(s)
| | - Daniel J-F Chinnapen
- Division of Gastroenterology, Boston Children's Hospital, Boston, Massachusetts 02155.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02155.,Department of Pediatrics, Harvard Digestive Diseases Center, Boston, Massachusetts 02155
| | - Wayne I Lencer
- Division of Gastroenterology, Boston Children's Hospital, Boston, Massachusetts 02155.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02155.,Department of Pediatrics, Harvard Digestive Diseases Center, Boston, Massachusetts 02155
| |
Collapse
|
13
|
Vogel GF, Janecke AR, Krainer IM, Gutleben K, Witting B, Mitton SG, Mansour S, Ballauff A, Roland JT, Engevik AC, Cutz E, Müller T, Goldenring JR, Huber LA, Hess MW. Abnormal Rab11-Rab8-vesicles cluster in enterocytes of patients with microvillus inclusion disease. Traffic 2017; 18:453-464. [PMID: 28407399 DOI: 10.1111/tra.12486] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/05/2017] [Accepted: 04/05/2017] [Indexed: 12/14/2022]
Abstract
Microvillus inclusion disease (MVID) is a congenital enteropathy characterized by accumulation of vesiculo-tubular endomembranes in the subapical cytoplasm of enterocytes, historically termed "secretory granules." However, neither their identity nor pathophysiological significance is well defined. Using immunoelectron microscopy and tomography, we studied biopsies from MVID patients (3× Myosin 5b mutations and 1× Syntaxin3 mutation) and compared them to controls and genome-edited CaCo2 cell models, harboring relevant mutations. Duodenal biopsies from 2 patients with novel Myosin 5b mutations and typical clinical symptoms showed unusual ultrastructural phenotypes: aberrant subapical vesicles and tubules were prominent in the enterocytes, though other histological hallmarks of MVID were almost absent (ectopic intra-/intercellular microvilli, brush border atrophy). We identified these enigmatic vesiculo-tubular organelles as Rab11-Rab8-positive recycling compartments of altered size, shape and location harboring the apical SNARE Syntaxin3, apical transporters sodium-hydrogen exchanger 3 (NHE3) and cystic fibrosis transmembrane conductance regulator. Our data strongly indicate that in MVID disrupted trafficking between cargo vesicles and the apical plasma membrane is the primary cause of a defect of epithelial polarity and subsequent facultative loss of brush border integrity, leading to malabsorption. Furthermore, they support the notion that mislocalization of transporters, such as NHE3 substantially contributes to the reported sodium loss diarrhea.
Collapse
Affiliation(s)
- Georg F Vogel
- Division of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria.,Division of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria.,Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas R Janecke
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Iris M Krainer
- Division of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria.,Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Karin Gutleben
- Division of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Witting
- Division of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Sahar Mansour
- Human Genetics Research Center, St. George's University of London, London, UK
| | | | - Joseph T Roland
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee.,Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee.,Departments of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Amy C Engevik
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee.,Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee.,Departments of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Ernest Cutz
- Division of Pathology, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Canada
| | - Thomas Müller
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - James R Goldenring
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee.,Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee.,Departments of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Lukas A Huber
- Division of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael W Hess
- Division of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
14
|
Lapierre LA, Manning EH, Mitchell KM, Caldwell CM, Goldenring JR. Interaction of phosphorylated Rab11-FIP2 with Eps15 regulates apical junction composition. Mol Biol Cell 2017; 28:1088-1100. [PMID: 28228550 PMCID: PMC5391185 DOI: 10.1091/mbc.e16-04-0214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 01/26/2017] [Accepted: 02/17/2017] [Indexed: 12/25/2022] Open
Abstract
MARK2 regulates the establishment of polarity in Madin-Darby canine kidney (MDCK) cells in part through phosphorylation of serine 227 of Rab11-FIP2. We identified Eps15 as an interacting partner of phospho-S227-Rab11-FIP2 (pS227-FIP2). During recovery from low calcium, Eps15 localized to the lateral membrane before pS227-FIP2 arrival. Later in recovery, Eps15 and pS227-FIP2 colocalized at the lateral membrane. In MDCK cells expressing the pseudophosphorylated FIP2 mutant FIP2(S227E), during recovery from low calcium, Eps15 was trapped and never localized to the lateral membrane. Mutation of any of the three NPF domains within GFP-FIP2(S227E) rescued Eps15 localization at the lateral membrane and reestablished single-lumen cyst formation in GFP-FIP2(S227E)-expressing cells in three-dimensional (3D) culture. Whereas expression of GFP-FIP2(S227E) induced the loss of E-cadherin and occludin, mutation of any of the NPF domains of GFP-FIP2(S227E) reestablished both proteins at the apical junctions. Knockdown of Eps15 altered the spatial and temporal localization of pS227-FIP2 and also elicited formation of multiple lumens in MDCK 3D cysts. Thus an interaction of Eps15 and pS227-FIP2 at the appropriate time and location in polarizing cells is necessary for proper establishment of epithelial polarity.
Collapse
Affiliation(s)
- Lynne A Lapierre
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232.,Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232.,Nashville VA Medical Center, Nashville, TN 37212
| | - Elizabeth H Manning
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232.,Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232.,Nashville VA Medical Center, Nashville, TN 37212
| | - Kenya M Mitchell
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232.,Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232.,Nashville VA Medical Center, Nashville, TN 37212
| | - Cathy M Caldwell
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232.,Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232.,Nashville VA Medical Center, Nashville, TN 37212
| | - James R Goldenring
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232 .,Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232.,Nashville VA Medical Center, Nashville, TN 37212.,Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232.,Vanderbilt Ingram Cancer Center, Nashville, TN 37232
| |
Collapse
|
15
|
Proteomics Screen Identifies Class I Rab11 Family Interacting Proteins as Key Regulators of Cytokinesis. Mol Cell Biol 2017; 37:MCB.00278-16. [PMID: 27872148 DOI: 10.1128/mcb.00278-16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 11/11/2016] [Indexed: 01/08/2023] Open
Abstract
The 14-3-3 protein family orchestrates a complex network of molecular interactions that regulates various biological processes. Owing to their role in regulating the cell cycle and protein trafficking, 14-3-3 proteins are prevalent in human diseases such as cancer, diabetes, and neurodegeneration. 14-3-3 proteins are expressed in all eukaryotic cells, suggesting that they mediate their biological functions through evolutionarily conserved protein interactions. To identify these core 14-3-3 client proteins, we used an affinity-based proteomics approach to characterize and compare the human and Drosophila 14-3-3 interactomes. Using this approach, we identified a group of Rab11 effector proteins, termed class I Rab11 family interacting proteins (Rab11-FIPs), or Rip11 in Drosophila We found that 14-3-3 binds to Rip11 in a phospho-dependent manner to ensure its proper subcellular distribution during cell division. Our results indicate that Rip11 plays an essential role in the regulation of cytokinesis and that this function requires its association with 14-3-3 but not with Rab11. Together, our results suggest an evolutionarily conserved role for 14-3-3 in controlling Rip11-dependent protein transport during cytokinesis.
Collapse
|
16
|
McRae R, Lapierre LA, Manning EH, Goldenring JR. Rab11-FIP1 phosphorylation by MARK2 regulates polarity in MDCK cells. CELLULAR LOGISTICS 2017; 7:e1271498. [PMID: 28396819 DOI: 10.1080/21592799.2016.1271498] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/30/2016] [Accepted: 12/06/2016] [Indexed: 10/20/2022]
Abstract
MARK2/Par1b/EMK1, a serine/threonine kinase, is required for correct apical/basolateral membrane polarization in epithelial cells. However, the specific substrates mediating MARK2 action are less well understood. We have now found that MARK2 phosphorylates Rab11-FIP1B/C at serine 234 in a consensus site similar to that previously identified in Rab11-FIP2. In MDCK cells undergoing repolarization after a calcium switch, antibodies specific for pS234-Rab11-FIP1 or pS227-Rab11-FIP2 demonstrate that the spatial and temporal activation of Rab11-FIP1 phosphorylation is distinct from that for Rab11-FIP2. Phosphorylation of Rab11-FIP1 persists through calcium switch and remains high after polarity has been reestablished whereas FIP2 phosphorylation is highest early in reestablishment of polarity but significantly reduced once polarity has been re-established. MARK2 colocalized with FIP1B/C/D and p(S234)-FIP1 in vivo. Overexpression of GFP-Rab11-FIP1C wildtype or non-phosphorylatable GFP-Rab11-FIP1C(S234A) induced two significant phenotypes following calcium switch. Overexpression of FIP1C wildtype and FIP1C(S234A) caused a psuedo-stratification of cells in early time points following calcium switch. At later time points most prominently observed in cells expressing FIP1C(S234A) a significant lateral lumen phenotype was observed, where F-actin-rich lateral lumens appeared demarcated by a ring of ZO1 and also containing ezrin, syntaxin 3 and podocalyxin. In contrast, p120 and E-Cadherin were excluded from the new apical surface at the lateral lumens and now localized to the new lateral surface oriented toward the media. GFP-FIP1C(S234A) localized to membranes deep to the lateral lumens, and immunostaining demonstrated the reorientation of the centrosome and the Golgi apparatus toward the lateral lumen. These results suggest that both Rab11-FIP1B/C and Rab11-FIP2 serve as critical substrates mediating aspects of MARK2 regulation of epithelial polarity.
Collapse
Affiliation(s)
- Rebecca McRae
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Lynne A Lapierre
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA; Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA; Nashville VA Medical Center, Nashville, TN, USA
| | - Elizabeth H Manning
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA; Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA; Nashville VA Medical Center, Nashville, TN, USA
| | - James R Goldenring
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA; Nashville VA Medical Center, Nashville, TN, USA; Vanderbilt Ingram Cancer Center, Nashville, TN, USA
| |
Collapse
|
17
|
Boulay PL, Mitchell L, Turpin J, Huot-Marchand JÉ, Lavoie C, Sanguin-Gendreau V, Jones L, Mitra S, Livingstone JM, Campbell S, Hallett M, Mills GB, Park M, Chodosh L, Strathdee D, Norman JC, Muller WJ. Rab11-FIP1C Is a Critical Negative Regulator in ErbB2-Mediated Mammary Tumor Progression. Cancer Res 2016; 76:2662-74. [PMID: 26933086 PMCID: PMC5070470 DOI: 10.1158/0008-5472.can-15-2782] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 02/05/2016] [Indexed: 02/06/2023]
Abstract
Rab coupling protein (FIP1C), an effector of the Rab11 GTPases, including Rab25, is amplified and overexpressed in 10% to 25% of primary breast cancers and correlates with poor clinical outcome. Rab25 is also frequently silenced in triple-negative breast cancer, suggesting its ability to function as either an oncogene or a tumor suppressor, depending on the breast cancer subtype. However, the pathobiologic role of FIP family members, such as FIP1C, in a tumor-specific setting remains elusive. In this study, we used ErbB2 mouse models of human breast cancer to investigate FIP1C function in tumorigenesis. Doxycycline-induced expression of FIP1C in the MMTV-ErbB2 mouse model resulted in delayed mammary tumor progression. Conversely, targeted deletion of FIP1C in the mammary epithelium of an ErbB2 model coexpressing Cre recombinase led to accelerated tumor onset. Genetic and biochemical characterization of these FIP1C-proficient and -deficient tumor models revealed that FIP1C regulated E-cadherin (CDH1) trafficking and ZONAB (YBX3) function in Cdk4-mediated cell-cycle progression. Furthermore, we demonstrate that FIP1C promoted lysosomal degradation of ErbB2. Consistent with our findings in the mouse, the expression of FIP1C was inversely correlated with ErbB2 levels in breast cancer patients. Taken together, our findings indicate that FIP1C acts as a tumor suppressor in the context of ErbB2-positive breast cancer and may be therapeutically exploited as an alternative strategy for targeting aberrant ErbB2 expression. Cancer Res; 76(9); 2662-74. ©2016 AACR.
Collapse
Affiliation(s)
- Pierre-Luc Boulay
- Department of Biochemistry, McGill University, Rosalind and Morris Goodman Cancer Research Montreal, Québec, Canada
| | - Louise Mitchell
- Integrin Cell Biology Cancer Research UK Beaston Institute, Glasgow, United Kingdom
| | - Jason Turpin
- Department of Biochemistry, McGill University, Rosalind and Morris Goodman Cancer Research Montreal, Québec, Canada
| | - Julie-Émilie Huot-Marchand
- Department of Biochemistry, McGill University, Rosalind and Morris Goodman Cancer Research Montreal, Québec, Canada
| | - Cynthia Lavoie
- Department of Biochemistry, McGill University, Rosalind and Morris Goodman Cancer Research Montreal, Québec, Canada
| | - Virginie Sanguin-Gendreau
- Department of Biochemistry, McGill University, Rosalind and Morris Goodman Cancer Research Montreal, Québec, Canada
| | - Laura Jones
- Department of Biochemistry, McGill University, Rosalind and Morris Goodman Cancer Research Montreal, Québec, Canada
| | - Shreya Mitra
- Department of System Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Julie M Livingstone
- Department of Biochemistry, McGill University, Rosalind and Morris Goodman Cancer Research Montreal, Québec, Canada
| | - Shirley Campbell
- Department of Pharmacology, University of Montreal, Québec, Canada
| | - Michael Hallett
- Department of Biochemistry, McGill University, Rosalind and Morris Goodman Cancer Research Montreal, Québec, Canada
| | - Gordon B Mills
- Department of System Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Morag Park
- Department of Biochemistry, McGill University, Rosalind and Morris Goodman Cancer Research Montreal, Québec, Canada
| | - Lewis Chodosh
- Cancer Biology Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Douglas Strathdee
- Integrin Cell Biology Cancer Research UK Beaston Institute, Glasgow, United Kingdom
| | - Jim C Norman
- Integrin Cell Biology Cancer Research UK Beaston Institute, Glasgow, United Kingdom
| | - William J Muller
- Department of Biochemistry, McGill University, Rosalind and Morris Goodman Cancer Research Montreal, Québec, Canada.
| |
Collapse
|
18
|
Weis VG, Knowles BC, Choi E, Goldstein AE, Williams JA, Manning EH, Roland JT, Lapierre LA, Goldenring JR. Loss of MYO5B in mice recapitulates Microvillus Inclusion Disease and reveals an apical trafficking pathway distinct to neonatal duodenum. Cell Mol Gastroenterol Hepatol 2016; 2:131-157. [PMID: 27019864 PMCID: PMC4806369 DOI: 10.1016/j.jcmgh.2015.11.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 11/25/2015] [Indexed: 12/10/2022]
Abstract
BACKGROUND AND AIMS Inactivating mutations in MYO5B cause severe neonatal diarrhea in Microvillus Inclusion Disease. Loss of active MYO5B causes the formation of pathognomonic inclusions and aberrations in brush border enzymes. METHODS We developed three mouse models of germline, constitutively intestinal targeted and inducible intestinal targeted deletion of MYO5B. The mice were evaluated for enterocyte cellular morphology. RESULTS Germline MYO5B KO mice showed early diarrhea and failure to thrive with evident microvillus inclusions and loss of apical transporters in the duodenum. IgG was present within inclusions. Apical transporters were lost and inclusions were present in the duodenum, but were nearly absent in the ileum. VillinCre;MYO5BF/F mice showed similar pathology and morphological changes in duodenal enterocytes. In contrast, when MYO5B KO was induced with tamoxifen treatment at 8 weeks of age, VillinCreERT2;MYO5BF/F mice developed severe diarrhea with loss of duodenal brush border enzymes, but few inclusions were observed in enterocytes. However, if tamoxifen is administered to 2-day-old VillinCreERT2;MYO5BF/F mice, prominent microvillus inclusions were observed. CONCLUSIONS The microvillus inclusions that develop after MYO5B loss reveal the presence of an unrecognized apical membrane trafficking pathway in neonatal duodenal enterocytes. However, the diarrheal pathology after MYO5B loss is due to deficits in transporter presentation at the apical membrane in duodenal enterocytes.
Collapse
Affiliation(s)
- Victoria G. Weis
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Byron C. Knowles
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Eunyoung Choi
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
- Nashville VA Medical Center, Nashville, Tennessee
| | - Anna E. Goldstein
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Janice A. Williams
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Elizabeth H. Manning
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
- Nashville VA Medical Center, Nashville, Tennessee
| | - Joseph T. Roland
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Lynne A. Lapierre
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
- Nashville VA Medical Center, Nashville, Tennessee
| | - James R. Goldenring
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Nashville VA Medical Center, Nashville, Tennessee
| |
Collapse
|
19
|
Schafer JC, McRae RE, Manning EH, Lapierre LA, Goldenring JR. Rab11-FIP1A regulates early trafficking into the recycling endosomes. Exp Cell Res 2016; 340:259-73. [PMID: 26790954 PMCID: PMC4744548 DOI: 10.1016/j.yexcr.2016.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 12/19/2015] [Accepted: 01/10/2016] [Indexed: 12/31/2022]
Abstract
The Rab11 family of small GTPases, along with the Rab11-family interacting proteins (Rab11-FIPs), are critical regulators of intracellular vesicle trafficking and recycling. We have identified a point mutation of Threonine-197 site to an Alanine in Rab11-FIP1A, which causes a dramatic dominant negative phenotype when expressed in HeLa cells. The normally perinuclear distribution of GFP-Rab11-FIP1A was condensed into a membranous cisternum with almost no GFP-Rab11-FIP1A(T197A) remaining outside of this central locus. Also, this condensed GFP-FIP1A(T197A) altered the distribution of proteins in the Rab11a recycling pathway including endogenous Rab11a, Rab11-FIP1C, and transferrin receptor (CD71). Furthermore, this condensed GFP-FIP1A(T197A)-containing structure exhibited little movement in live HeLa cells. Expression of GFP-FIP1A(T197A) caused a strong blockade of transferrin recycling. Treatment of cells expressing GFP-FIP1A(T197A) with nocodazole did not disperse the Rab11a-containing recycling system. We also found that Rab5 and EEA1 were accumulated in membranes by GFP-Rab11-FIP1A but Rab4 was unaffected, suggesting that a direct pathway may exist from early endosomes into the Rab11a-containing recycling system. Our study of a potent inhibitory trafficking mutation in Rab11-FIP1A shows that Rab11-FIP1A associates with and regulates trafficking at an early step in the process of membrane recycling.
Collapse
Affiliation(s)
- Jenny C Schafer
- Departments of Surgery, Nashville, TN, USA; Epithelial Biology Center, Nashville, TN, USA
| | - Rebecca E McRae
- Departments of Surgery, Nashville, TN, USA; Cell & Developmental Biology, Nashville, TN, USA; Epithelial Biology Center, Nashville, TN, USA
| | - Elizabeth H Manning
- Departments of Surgery, Nashville, TN, USA; Epithelial Biology Center, Nashville, TN, USA
| | - Lynne A Lapierre
- Departments of Surgery, Nashville, TN, USA; Epithelial Biology Center, Nashville, TN, USA
| | - James R Goldenring
- Departments of Surgery, Nashville, TN, USA; Cell & Developmental Biology, Nashville, TN, USA; Epithelial Biology Center, Nashville, TN, USA; Vanderbilt University School of Medicine and the Nashville VA Medical Center, Nashville, TN, USA.
| |
Collapse
|
20
|
Zhao Z, Sagare AP, Ma Q, Halliday MR, Kong P, Kisler K, Winkler EA, Ramanathan A, Kanekiyo T, Bu G, Owens NC, Rege SV, Si G, Ahuja A, Zhu D, Miller CA, Schneider JA, Maeda M, Maeda T, Sugawara T, Ichida JK, Zlokovic BV. Central role for PICALM in amyloid-β blood-brain barrier transcytosis and clearance. Nat Neurosci 2015; 18:978-87. [PMID: 26005850 PMCID: PMC4482781 DOI: 10.1038/nn.4025] [Citation(s) in RCA: 324] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 04/21/2015] [Indexed: 12/11/2022]
Abstract
PICALM is a highly validated genetic risk factor for Alzheimer's disease (AD). We found that reduced expression of PICALM in AD and murine brain endothelium correlated with amyloid-β (Aβ) pathology and cognitive impairment. Moreover, Picalm deficiency diminished Aβ clearance across the murine blood-brain barrier (BBB) and accelerated Aβ pathology in a manner that was reversible by endothelial PICALM re-expression. Using human brain endothelial monolayers, we found that PICALM regulated PICALM/clathrin-dependent internalization of Aβ bound to the low density lipoprotein receptor related protein-1, a key Aβ clearance receptor, and guided Aβ trafficking to Rab5 and Rab11, leading to Aβ endothelial transcytosis and clearance. PICALM levels and Aβ clearance were reduced in AD-derived endothelial monolayers, which was reversible by adenoviral-mediated PICALM transfer. Inducible pluripotent stem cell-derived human endothelial cells carrying the rs3851179 protective allele exhibited higher PICALM levels and enhanced Aβ clearance. Thus, PICALM regulates Aβ BBB transcytosis and clearance, which has implications for Aβ brain homeostasis and clearance therapy.
Collapse
Affiliation(s)
- Zhen Zhao
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Abhay P. Sagare
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Qingyi Ma
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Matthew R. Halliday
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Pan Kong
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kassandra Kisler
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ethan A. Winkler
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Anita Ramanathan
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Nelly Chuqui Owens
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Sanket V. Rege
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Gabriel Si
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ashim Ahuja
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Donghui Zhu
- Department of Chemical, Biological and Bio–Engineering, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA
| | - Carol A. Miller
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Julie A. Schneider
- Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Manami Maeda
- Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Takahiro Maeda
- Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tohru Sugawara
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, 1425 San Pablo Street, BCC 307, Los Angeles, CA 90089, USA
| | - Justin K. Ichida
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, 1425 San Pablo Street, BCC 307, Los Angeles, CA 90089, USA
| | - Berislav V. Zlokovic
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
21
|
Goldenring JR. Recycling endosomes. Curr Opin Cell Biol 2015; 35:117-22. [PMID: 26022676 DOI: 10.1016/j.ceb.2015.04.018] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/29/2015] [Accepted: 04/30/2015] [Indexed: 12/13/2022]
Abstract
The endosomal membrane recycling system represents a dynamic conduit for sorting and re-exporting internalized membrane constituents. The recycling system is composed of multiple tubulovesicular recycling pathways that likely confer distinct trafficking pathways for individual cargoes. In addition, elements of the recycling system are responsible for assembly and maintenance of apical membrane specializations including primary cilia and apical microvilli. The existence of multiple intersecting and diverging recycling tracks likely accounts for specificity in plasma membrane recycling trafficking.
Collapse
Affiliation(s)
- James R Goldenring
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; The Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA; The Nashville VA Medical Center, Nashville, TN, USA.
| |
Collapse
|
22
|
Proteomic analysis of proteins surrounding occludin and claudin-4 reveals their proximity to signaling and trafficking networks. PLoS One 2015; 10:e0117074. [PMID: 25789658 PMCID: PMC4366163 DOI: 10.1371/journal.pone.0117074] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/18/2014] [Indexed: 01/15/2023] Open
Abstract
Tight junctions are complex membrane structures that regulate paracellular movement of material across epithelia and play a role in cell polarity, signaling and cytoskeletal organization. In order to expand knowledge of the tight junction proteome, we used biotin ligase (BioID) fused to occludin and claudin-4 to biotinylate their proximal proteins in cultured MDCK II epithelial cells. We then purified the biotinylated proteins on streptavidin resin and identified them by mass spectrometry. Proteins were ranked by relative abundance of recovery by mass spectrometry, placed in functional categories, and compared not only among the N- and C- termini of occludin and the N-terminus of claudin-4, but also with our published inventory of proteins proximal to the adherens junction protein E-cadherin and the tight junction protein ZO-1. When proteomic results were analyzed, the relative distribution among functional categories was similar between occludin and claudin-4 proximal proteins. Apart from already known tight junction- proteins, occludin and claudin-4 proximal proteins were enriched in signaling and trafficking proteins, especially endocytic trafficking proteins. However there were significant differences in the specific proteins comprising the functional categories near each of the tagging proteins, revealing spatial compartmentalization within the junction complex. Taken together, these results expand the inventory of known and unknown proteins at the tight junction to inform future studies of the organization and physiology of this complex structure.
Collapse
|
23
|
Knowles BC, Weis VG, Yu S, Roland JT, Williams JA, Alvarado GS, Lapierre LA, Shub MD, Gao N, Goldenring JR. Rab11a regulates syntaxin 3 localization and microvillus assembly in enterocytes. J Cell Sci 2015; 128:1617-26. [PMID: 25673875 DOI: 10.1242/jcs.163303] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 02/02/2015] [Indexed: 02/02/2023] Open
Abstract
Rab11a is a key component of the apical recycling endosome that aids in the trafficking of proteins to the luminal surface in polarized epithelial cells. Utilizing conditional Rab11a-knockout specific to intestinal epithelial cells, and human colonic epithelial CaCo2-BBE cells with stable Rab11a knockdown, we examined the molecular and pathological impact of Rab11a deficiency on the establishment of apical cell polarity and microvillus morphogenesis. We demonstrate that loss of Rab11a induced alterations in enterocyte polarity, shortened microvillar length and affected the formation of microvilli along the lateral membranes. Rab11a deficiency in enterocytes altered the apical localization of syntaxin 3. These data affirm the role of Rab11a in apical membrane trafficking and the maintenance of apical microvilli in enterocytes.
Collapse
Affiliation(s)
- Byron C Knowles
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37235, USA Department of Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37235, USA
| | - Victoria G Weis
- Department of Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37235, USA Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN 37235, USA
| | - Shiyan Yu
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Joseph T Roland
- Department of Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37235, USA Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN 37235, USA
| | - Janice A Williams
- Vanderbilt Ingraham Cancer Center: Cell Imaging Shared Resource, Vanderbilt University School of Medicine, Nashville, TN 37235, USA
| | - Gabriela S Alvarado
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37235, USA Department of Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37235, USA
| | - Lynne A Lapierre
- Department of Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37235, USA Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN 37235, USA
| | - Mitchell D Shub
- Phoenix Children's Hospital and the Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA Rutgers Cancer Institute of New Jersey, Piscataway, NJ 08903, USA
| | - James R Goldenring
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37235, USA Department of Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37235, USA Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN 37235, USA
| |
Collapse
|
24
|
Knowles BC, Roland JT, Krishnan M, Tyska MJ, Lapierre LA, Dickman PS, Goldenring JR, Shub MD. Myosin Vb uncoupling from RAB8A and RAB11A elicits microvillus inclusion disease. J Clin Invest 2014; 124:2947-62. [PMID: 24892806 DOI: 10.1172/jci71651] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 04/17/2014] [Indexed: 12/14/2022] Open
Abstract
Microvillus inclusion disease (MVID) is a severe form of congenital diarrhea that arises from inactivating mutations in the gene encoding myosin Vb (MYO5B). We have examined the association of mutations in MYO5B and disruption of microvillar assembly and polarity in enterocytes. Stable MYO5B knockdown (MYO5B-KD) in CaCo2-BBE cells elicited loss of microvilli, alterations in junctional claudins, and disruption of apical and basolateral trafficking; however, no microvillus inclusions were observed in MYO5B-KD cells. Expression of WT MYO5B in MYO5B-KD cells restored microvilli; however, expression of MYO5B-P660L, a MVID-associated mutation found within Navajo populations, did not rescue the MYO5B-KD phenotype but induced formation of microvillus inclusions. Microvilli establishment required interaction between RAB8A and MYO5B, while loss of the interaction between RAB11A and MYO5B induced microvillus inclusions. Using surface biotinylation and dual immunofluorescence staining in MYO5B-KD cells expressing mutant forms of MYO5B, we observed that early microvillus inclusions were positive for the sorting marker SNX18 and derived from apical membrane internalization. In patients with MVID, MYO5B-P660L results in global changes in polarity at the villus tips that could account for deficits in apical absorption, loss of microvilli, aberrant junctions, and losses in transcellular ion transport pathways, likely leading to the MVID clinical phenotype of neonatal secretory diarrhea.
Collapse
|
25
|
Lu R, Johnson DL, Stewart L, Waite K, Elliott D, Wilson JM. Rab14 regulation of claudin-2 trafficking modulates epithelial permeability and lumen morphogenesis. Mol Biol Cell 2014; 25:1744-54. [PMID: 24694596 PMCID: PMC4038501 DOI: 10.1091/mbc.e13-12-0724] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Epithelial permeability is regulated by targeted insertion and recycling of tight junction proteins. Rab14 regulates the lysosomal targeting of the leaky claudin, claudin-2, and depletion of Rab14 results in increased transepithelial resistance and aberrant morphogenesis in three-dimensional culture. Regulation of epithelial barrier function requires targeted insertion of tight junction proteins that have distinct selectively permeable characteristics. The insertion of newly synthesized proteins and recycling of internalized tight junction components control both polarity and junction function. Here we show that the small GTPase Rab14 regulates tight junction structure. In Madin–Darby canine kidney (MDCK) II cells, Rab14 colocalizes with junctional proteins, and knockdown of Rab14 results in increased transepithelial resistance. In cells without Rab14, there are small changes in the trafficking of claudin-1 and occludin. In addition, there is substantial depletion of the leaky claudin, claudin-2, but not other tight junction components. The loss of claudin-2 is complemented by inhibition of lysosomal function, suggesting that Rab14 sorts claudin-2 out of the lysosome-directed pathway. MDCK I cells lack claudin-2 endogenously, and knockdown of Rab14 in these cells does not result in a change in transepithelial resistance, suggesting that the effect is specific to claudin-2 trafficking. Furthermore, leaky claudins have been shown to be required for epithelial morphogenesis, and knockdown of Rab14 results in failure to form normal single-lumen cysts in three-dimensional culture. These results implicate Rab14 in specialized trafficking of claudin-2 from the recycling endosome.
Collapse
Affiliation(s)
- Ruifeng Lu
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ 85724
| | - Debra L Johnson
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ 85724
| | - Lorraine Stewart
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ 85724
| | - Kelsey Waite
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ 85724
| | - David Elliott
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ 85724
| | - Jean M Wilson
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ 85724
| |
Collapse
|
26
|
Schafer JC, Baetz NW, Lapierre LA, McRae RE, Roland JT, Goldenring JR. Rab11-FIP2 interaction with MYO5B regulates movement of Rab11a-containing recycling vesicles. Traffic 2014; 15:292-308. [PMID: 24372966 DOI: 10.1111/tra.12146] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 12/17/2013] [Accepted: 12/24/2013] [Indexed: 12/28/2022]
Abstract
A tripartite association of Rab11a with both Rab11-FIP2 and MYO5B regulates recycling endosome trafficking. We sought to define the intermolecular interactions required between Rab11-FIP2 and MYO5B. Using a random mutagenesis strategy, we identified point mutations at S229P or G233E in Rab11-FIP2 that caused loss of interaction with MYO5B in yeast two-hybrid assays as well as loss of interaction of Rab11-FIP2(129-356) with MYO5B tail when expressed in HeLa cells. Single mutations or the double S229P/G233E mutation failed to alter the association of full-length Rab11-FIP2 with MYO5B tail in HeLa cells. While EGFP-Rab11-FIP2 wild type colocalized with endogenous MYO5B staining in MDCK cells, EGFP-Rab11-FIP2(S229P/G233E) showed a significant decrease in localization with endogenous MYO5B. Analysis of Rab11a-containing vesicle movement in live HeLa cells demonstrated that when the MYO5B/Rab11-FIP2 association is perturbed by mutation or by Rab11-FIP2 knockdown, vesicle movement is increased in both speed and track length, consistent with an impairment of MYO5B tethering at the cytoskeleton. These results support a critical role for the interaction of MYO5B with Rab11-FIP2 in stabilizing the functional complex with Rab11a, which regulates dynamic movements of membrane recycling vesicles.
Collapse
Affiliation(s)
- Jenny C Schafer
- Section of Surgical Sciences and the Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | | | | | | |
Collapse
|
27
|
Lall P, Horgan CP, Oda S, Franklin E, Sultana A, Hanscom SR, McCaffrey MW, Khan AR. Structural and functional analysis of FIP2 binding to the endosome-localised Rab25 GTPase. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2679-90. [PMID: 24056041 DOI: 10.1016/j.bbapap.2013.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 09/08/2013] [Accepted: 09/12/2013] [Indexed: 11/30/2022]
Abstract
Rab small GTPases are the master regulators of intracellular trafficking in eukaryotes. They mediate spatial and temporal recruitment of effector proteins to distinct cellular compartments through GTP-induced changes in their conformation. Despite numerous structural studies, the molecular basis for Rab/effector specificity and subsequent biological activity remains poorly understood. Rab25, also known as Rab11c, which is epithelial-specific, has been heavily implicated in ovarian cancer development and independently appears to act as a tumour suppressor in the context of a distinct subset of carcinomas. Here, we show that Rab25 associates with FIP2 and can recruit this effector protein to endosomal membranes. We report the crystal structure of Rab25 in complex with the C-terminal region of FIP2, which consists of a central dimeric FIP2 coiled-coil that mediates a heterotetrameric Rab25-(FIP2)2-Rab25 complex. Thermodynamic analyses show that, despite a relatively conserved interface, FIP2 binds to Rab25 with an approximate 3-fold weaker affinity than to Rab11a. Reduced affinity is mainly associated with lower enthalpic gains for Rab25:FIP2 complex formation, and can be attributed to subtle differences in the conformations of switch 1 and switch 2. These cellular, structural and thermodynamic studies provide insight into the Rab11/Rab25 subfamily of small GTPases that regulate endosomal trafficking pathways in eukaryotes.
Collapse
Affiliation(s)
- Patrick Lall
- School of Biochemistry and Immunology, Trinity College, Dublin 2, Ireland
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Messenger SW, Thomas DDH, Falkowski MA, Byrne JA, Gorelick FS, Groblewski GE. Tumor protein D52 controls trafficking of an apical endolysosomal secretory pathway in pancreatic acinar cells. Am J Physiol Gastrointest Liver Physiol 2013; 305:G439-52. [PMID: 23868405 PMCID: PMC3761242 DOI: 10.1152/ajpgi.00143.2013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 07/12/2013] [Indexed: 01/31/2023]
Abstract
Zymogen granule (ZG) formation in acinar cells involves zymogen cargo sorting from trans-Golgi into immature secretory granules (ISGs). ISG maturation progresses by removal of lysosomal membrane and select content proteins, which enter endosomal intermediates prior to their apical exocytosis. Constitutive and stimulated secretion through this mechanism is termed the constitutive-like and minor-regulated pathways, respectively. However, the molecular components that control membrane trafficking within these endosomal compartments are largely unknown. We show that tumor protein D52 is highly expressed in endosomal compartments following pancreatic acinar cell stimulation and regulates apical exocytosis of an apically directed endolysosomal compartment. Secretion from the endolysosomal compartment was detected by cell-surface antigen labeling of lysosome-associated membrane protein LAMP1, which is absent from ZGs, and had incomplete overlap with surface labeling of synaptotagmin 1, a marker of ZG exocytosis. Although culturing (16-18 h) of isolated acinar cells is accompanied by a loss of secretory responsiveness, the levels of SNARE proteins necessary for ZG exocytosis were preserved. However, levels of endolysosomal proteins D52, EEA1, Rab5, and LAMP1 markedly decreased with culture. When D52 levels were restored by adenoviral delivery, the levels of these regulatory proteins and secretion of both LAMP1 (endolysosomal) and amylase was strongly enhanced. These secretory effects were absent in alanine and aspartate substitutions of serine 136, the major D52 phosphorylation site, and were inhibited by brefeldin A, which does not directly affect the ZG compartment. Our results indicate that D52 directly regulates apical endolysosomal secretion and are consistent with previous studies, suggesting that this pathway indirectly regulates ZG secretion of digestive enzymes.
Collapse
Affiliation(s)
- Scott W Messenger
- Univ. of Wisconsin, Dept. of Nutritional Sciences, 1415 Linden Dr., Madison, WI 53706.
| | | | | | | | | | | |
Collapse
|
29
|
Krishnan M, Lapierre LA, Knowles BC, Goldenring JR. Rab25 regulates integrin expression in polarized colonic epithelial cells. Mol Biol Cell 2013; 24:818-31. [PMID: 23345591 PMCID: PMC3596252 DOI: 10.1091/mbc.e12-10-0745] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Rab25 is a tumor suppressor in the colon, but the mechanisms underlying the influence of Rab25 on polarity are unknown. Findings on changes in polarity in Caco2-BBE cells with knockdown and rescue of Rab25 expression indicate that Rab25 regulates integrin gene expression mediated by ETV4. Rab25 is a tumor suppressor for colon cancer in humans and mice. To identify elements of intestinal polarity regulated by Rab25, we developed Caco2-BBE cell lines stably expressing short hairpin RNA for Rab25 and lines rescuing Rab25 knockdown with reexpression of rabbit Rab25. Rab25 knockdown decreased α2-, α5-, and β1-integrin expression. We observed colocalization and direct association of Rab25 with α5β1-integrins. Rab25 knockdown also up-regulated claudin-1 expression, increased transepithelial resistance, and increased invasive behavior. Rab25-knockdown cells showed disorganized brush border microvilli with decreases in villin expression. All of these changes were reversed by reintroduction of rabbit Rab25. Rab25 knockdown altered the expression of 29 gene transcripts, including the loss of α5-integrin transcripts. Rab25 loss decreased expression of one transcription factor, ETV4, and overexpression of ETV4 in Rab25-knockdown cells reversed losses of α5β1-integrin. The results suggest that Rab25 controls intestinal cell polarity through the regulation of gene expression.
Collapse
Affiliation(s)
- Moorthy Krishnan
- Section of Surgical Sciences and the Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | |
Collapse
|