1
|
Wang H, Yan C, Ye H. RNF126 Promotes Ovarian Cancer Progression by Reprogramming Lipid Metabolism Through Degradation of ACAP2. Biochem Genet 2025:10.1007/s10528-025-11107-1. [PMID: 40251363 DOI: 10.1007/s10528-025-11107-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 04/12/2025] [Indexed: 04/20/2025]
Abstract
Ovarian cancer (OC) primarily arises from heterogeneous malignant epithelial tissue in the ovary, fallopian tube, or peritoneum. Ubiquitin ligase Ring-finger protein 126 (RNF126) was aberrantly expressed in OC. However, its molecular mechanism is unknown. This study investigates the role and mechanism of RNF126 in regulating ArfGAP with coiled-coil, ankyrin repeat, and PH domains 2 (ACAP2) during OC progression. RT-qPCR and Western blot (WB) were used to assess the expression of RNF126, ACAP2, and lipid synthesis-related genes in OC tissues and cells. The proliferation and migration abilities of OC cells were detected by CCK-8 and Transwell assays. Nile red staining was used to detect the lipid accumulation. The interaction between RNF126 and ACAP2 in OC cells was detected using co-immunoprecipitation (Co-IP). The stability of the ACAP2 protein was analyzed using the cycloheximide (CHX) assay. The effect of RNF126 on tumor growth and metastasis in vivo was investigated by detecting tumor volume and size as well as the number of lung nodules. The expression of RNF126 was upregulated in OC tissues and cells, and RNF126 silencing significantly inhibited the proliferation, migration, and lipid accumulation of OC cells. Mechanistically, ACAP2 was identified as a ubiquitination substrate of RNF126, and its expression was negatively regulated by RNF126. Furthermore, RNF126 promoted OC progression both in vitro and in vivo by suppressing ACAP2 protein expression. RNF126 promotes ovarian cancer progression by reprogramming lipid metabolism via degrading ACAP2.
Collapse
Affiliation(s)
- Hao Wang
- Department of Gynecology and Obstetrics, The First College of Clinical Medical Science, China Three Gorges University, No. 183, Yiling Avenue, Yichang, 443000, Hubei, China
| | - Chao Yan
- Department of Orthopedics, College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
| | - Hong Ye
- Department of Gynecology and Obstetrics, The First College of Clinical Medical Science, China Three Gorges University, No. 183, Yiling Avenue, Yichang, 443000, Hubei, China.
| |
Collapse
|
2
|
Sun X, Li Y, He Y, Cheng L, Wang L, Wei J, Chen J, Du L, Shen Z, Xie Y, Midgley AC, Jiang W, Yoshida S. Aberrant expression of GTPase-activating protein ARAP1 triggers circular dorsal ruffles associated with malignancy in hepatocellular carcinoma Hep3B cells. Cell Commun Signal 2025; 23:75. [PMID: 39934854 PMCID: PMC11816549 DOI: 10.1186/s12964-025-02084-4] [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: 10/25/2024] [Accepted: 02/05/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Circular dorsal ruffles (CDRs) are large and rounded membrane ruffles that function as precursors of macropinocytosis. We recently reported that CDRs form in Hep3B hepatocellular carcinoma (HCC) cells, but not in Huh7 and HepG2 HCC cells or LO2 cells, suggesting that an unknown molecular mechanism implicates CDRs in Hep3B malignancy through macropinocytosis uptake of excessive extracellular nutrients. In this study, we investigated the cellular role and the mechanism of CDRs in Hep3B cells by focusing on the GTPase-activating protein ARAP1. METHODS ARAP1 knock-out (KO) cells were generated. Confocal microscopy and high-resolution scanning electron microscopy (SEM) were used for identification of the target proteins and structure analysis, respectively. Proteasome inhibitor MG132, mitochondrial function inhibitor CCCP, ARF1 inhibitor Golgicide A, and macropinocytosis inhibitor EIPA were used to investigate the molecular mechanism. Cell proliferation and Transwell migration/invasion assays were used to investigate the role of ARAP1 in cellular malignancy. RESULTS ARAP1 was localized to CDRs, which had reduced size following ARAP1 KO. CDRs comprised small vertical lamellipodia, the expression pattern of which was disrupted in ARAP1 KO cells. Extracellular solute uptake, rate of cell growth, and malignant potential were attenuated in KO cells. ARAP1 was also localized to mitochondria in Hep3B cells but not in the control cell lines. Mitochondrial fission protein was increased in KO cells. CCCP treatment blocked CDRs in Hep3B cells but not in controls. Surprisingly, ARAP1 expression level in Hep3B cells was lower than in Huh7, HepG2, and LO2 cells. MG132 treatment increased the ARAP1 levels in Hep3B cells, but not in Huh7 cells, revealing that ARAP1 is actively degraded in Hep3B cells. CONCLUSIONS These results strongly suggest that the aberrant expression of ARAP1 in Hep3B cells modulates CDRs via mitochondrial function, thereby resulting in excess uptake of nutrients as an initial event in cancer development. Based on these findings, we propose that the molecular mechanisms underlying the formation of CDRs, focusing on ARAP1, may serve as an effective therapeutic target in some types of HCC and cancers.
Collapse
Affiliation(s)
- Xiaowei Sun
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, China
| | - Yanan Li
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, China
| | - Yuxin He
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, China
| | - Longjiao Cheng
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, China
| | - Li Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, China
| | - Jinzi Wei
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, China
| | - Jianan Chen
- Organ Transplant Department, School of Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Linxuan Du
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, China
| | - Zhongyang Shen
- Organ Transplant Department, School of Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
- Research Institute of Transplant Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory for Organ Transplantation, Tianjin, China
| | - Yan Xie
- Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Tianjin First Center Hospital, Tianjin, 300384, China.
- Liver Transplantation Department, Tianjin First Center Hospital, Tianjin, China.
| | - Adam C Midgley
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, China.
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Wentao Jiang
- Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Tianjin First Center Hospital, Tianjin, 300384, China.
- Liver Transplantation Department, Tianjin First Center Hospital, Tianjin, China.
| | - Sei Yoshida
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Frontiers Science Center for Cell Responses, Nankai University, Tianjin, China.
- Research Institute of Transplant Medicine, Nankai University, Tianjin, China.
- Nankai International Advanced Research Institute, Shenzhen, China.
- Nankai University College of Life Sciences, Tianjin, 300071, China.
| |
Collapse
|
3
|
Wee Y, Wang J, Wilson EC, Rich CP, Rogers A, Tong Z, DeGroot E, Gopal YNV, Davies MA, Ekiz HA, Tay JKH, Stubben C, Boucher KM, Oviedo JM, Fairfax KC, Williams MA, Holmen SL, Wolff RK, Grossmann AH. Tumour-intrinsic endomembrane trafficking by ARF6 shapes an immunosuppressive microenvironment that drives melanomagenesis and response to checkpoint blockade therapy. Nat Commun 2024; 15:6613. [PMID: 39098861 PMCID: PMC11298541 DOI: 10.1038/s41467-024-50881-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 07/24/2024] [Indexed: 08/06/2024] Open
Abstract
Tumour-host immune interactions lead to complex changes in the tumour microenvironment (TME), impacting progression, metastasis and response to therapy. While it is clear that cancer cells can have the capacity to alter immune landscapes, our understanding of this process is incomplete. Herein we show that endocytic trafficking at the plasma membrane, mediated by the small GTPase ARF6, enables melanoma cells to impose an immunosuppressive TME that accelerates tumour development. This ARF6-dependent TME is vulnerable to immune checkpoint blockade therapy (ICB) but in murine melanoma, loss of Arf6 causes resistance to ICB. Likewise, downregulation of ARF6 in patient tumours correlates with inferior overall survival after ICB. Mechanistically, these phenotypes are at least partially explained by ARF6-dependent recycling, which controls plasma membrane density of the interferon-gamma receptor. Collectively, our findings reveal the importance of endomembrane trafficking in outfitting tumour cells with the ability to shape their immune microenvironment and respond to immunotherapy.
Collapse
Affiliation(s)
- Yinshen Wee
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- School of Dentistry, Taipei Medical University, Taipei, Taiwan
| | - Junhua Wang
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- Department of Oncologic Sciences, University of Utah, Salt Lake City, UT, USA
| | - Emily C Wilson
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Coulson P Rich
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Aaron Rogers
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Zongzhong Tong
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Evelyn DeGroot
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Y N Vashisht Gopal
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - H Atakan Ekiz
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Gulbahce, Urla, Izmir, Turkey
| | - Joshua K H Tay
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Chris Stubben
- Bioinformatics Shared Resource, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Kenneth M Boucher
- Cancer Biostatistics Shared Resource, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Juan M Oviedo
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Keke C Fairfax
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Matthew A Williams
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Sheri L Holmen
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- Department of Oncologic Sciences, University of Utah, Salt Lake City, UT, USA
- Department of Surgery, University of Utah, Salt Lake City, UT, USA
| | - Roger K Wolff
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Allie H Grossmann
- Department of Pathology, University of Utah, Salt Lake City, UT, USA.
- Huntsman Cancer Institute, Salt Lake City, UT, USA.
- Department of Oncologic Sciences, University of Utah, Salt Lake City, UT, USA.
- Providence Cancer Institute of Oregon, Earle A. Chiles Research Institute, Portland, OR, USA.
| |
Collapse
|
4
|
O'Donoghue L, Smolenski A. Roles of G proteins and their GTPase-activating proteins in platelets. Biosci Rep 2024; 44:BSR20231420. [PMID: 38808367 PMCID: PMC11139668 DOI: 10.1042/bsr20231420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/30/2024] Open
Abstract
Platelets are small anucleate blood cells supporting vascular function. They circulate in a quiescent state monitoring the vasculature for injuries. Platelets adhere to injury sites and can be rapidly activated to secrete granules and to form platelet/platelet aggregates. These responses are controlled by signalling networks that include G proteins and their regulatory guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). Recent proteomics studies have revealed the complete spectrum of G proteins, GEFs, and GAPs present in platelets. Some of these proteins are specific for platelets and very few have been characterised in detail. GEFs and GAPs play a major role in setting local levels of active GTP-bound G proteins in response to activating and inhibitory signals encountered by platelets. Thus, GEFs and GAPs are highly regulated themselves and appear to integrate G protein regulation with other cellular processes. This review focuses on GAPs of small G proteins of the Arf, Rab, Ras, and Rho families, as well as of heterotrimeric G proteins found in platelets.
Collapse
Affiliation(s)
- Lorna O'Donoghue
- UCD School of Medicine, University College Dublin, UCD Conway Institute, Belfield, Dublin 4, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green 123, Dublin 2, Ireland
| | - Albert Smolenski
- UCD School of Medicine, University College Dublin, UCD Conway Institute, Belfield, Dublin 4, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green 123, Dublin 2, Ireland
| |
Collapse
|
5
|
Sun X, Li Y, He Y, Cheng L, Wei J, Du L, Shen Z, Yoshida S. GTPase-activating protein ARAP1 regulates circular dorsal ruffles as a nutrient uptake mechanism in the Hep3B hepatocellular carcinoma cell line. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.31.573800. [PMID: 38260345 PMCID: PMC10802275 DOI: 10.1101/2023.12.31.573800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Circular dorsal ruffles (CDRs), large-scale rounded membrane ruffles, function as precursors of macropinocytosis. We recently reported that CDRs are exposed in the Hep3B hepatocellular carcinoma cell line, while not in other hepatocellular carcinoma cell lines, indicating that the CDRs in Hep3B are associated with malignant potential. In this study, we investigated the cellular function of CDRs in Hep3B cells by focusing on the molecular mechanisms of the GTPase-activating protein ARAP1. ARAP1 was localized to the CDRs, the sizes of which were reduced by deletion of this protein. High-resolution scanning electron micrographs revealed that CDRs comprise small vertical lamellipodia, the expression pattern of which was disrupted in ARAP1 KO cells. Extracellular solute uptake, rate of cell growth, and malignant potential were attenuated in the KO cells. ARAP1 is also localized in Hep3B cell mitochondria, although not in those of the Huh7 hepatocellular carcinoma cell line. On the basis of these findings, we propose that the aberrant expression of ARAP1 in Hep3B cells modulates CDRs, thereby resulting in an excess uptake of nutrients as an initial event in cancer development. SUMMARY STATEMENT ARAP1 regulates circular dorsal ruffles (CDRs) in the Hep3B HCC cell line and deletion of this protein attenuates malignant potential, thereby indicating the involvement of CDRs in cancer development.
Collapse
|
6
|
Wee Y, Wang J, Wilson EC, Rich CP, Rogers A, Tong Z, DeGroot E, Gopal YV, Davies MA, Ekiz HA, Tay JK, Stubben C, Boucher KM, Oviedo JM, Fairfax KC, Williams MA, Holmen SL, Wolff RK, Grossmann AH. ARF6-dependent endocytic trafficking of the Interferon-γ receptor drives adaptive immune resistance in cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.560199. [PMID: 37873189 PMCID: PMC10592860 DOI: 10.1101/2023.09.29.560199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Adaptive immune resistance (AIR) is a protective process used by cancer to escape elimination by CD8+ T cells. Inhibition of immune checkpoints PD-1 and CTLA-4 specifically target Interferon-gamma (IFNγ)-driven AIR. AIR begins at the plasma membrane where tumor cell-intrinsic cytokine signaling is initiated. Thus, plasma membrane remodeling by endomembrane trafficking could regulate AIR. Herein we report that the trafficking protein ADP-Ribosylation Factor 6 (ARF6) is critical for IFNγ-driven AIR. ARF6 prevents transport of the receptor to the lysosome, augmenting IFNγR expression, tumor intrinsic IFNγ signaling and downstream expression of immunosuppressive genes. In murine melanoma, loss of ARF6 causes resistance to immune checkpoint blockade (ICB). Likewise, low expression of ARF6 in patient tumors correlates with inferior outcomes with ICB. Our data provide new mechanistic insights into tumor immune escape, defined by ARF6-dependent AIR, and support that ARF6-dependent endomembrane trafficking of the IFNγ receptor influences outcomes of ICB.
Collapse
Affiliation(s)
- Yinshen Wee
- Department of Pathology, University of Utah, Salt Lake City, Utah
- Huntsman Cancer Institute, Salt Lake City, Utah
- These authors contributed equally
- current contact information: School of Dentistry, Taipei Medical University, Taiwan
| | - Junhua Wang
- Department of Pathology, University of Utah, Salt Lake City, Utah
- Huntsman Cancer Institute, Salt Lake City, Utah
- These authors contributed equally
| | - Emily C. Wilson
- Department of Pathology, University of Utah, Salt Lake City, Utah
- Huntsman Cancer Institute, Salt Lake City, Utah
| | - Coulson P. Rich
- Department of Pathology, University of Utah, Salt Lake City, Utah
- Huntsman Cancer Institute, Salt Lake City, Utah
| | - Aaron Rogers
- Department of Pathology, University of Utah, Salt Lake City, Utah
- Huntsman Cancer Institute, Salt Lake City, Utah
| | - Zongzhong Tong
- Department of Pathology, University of Utah, Salt Lake City, Utah
| | - Evelyn DeGroot
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Y.N. Vashisht Gopal
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael A. Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - H. Atakan Ekiz
- Department of Molecular Biology and Genetics, Izmir institute of Technology, Gulbahce, Urla, 35430, Izmir, Turkey
| | - Joshua K.H. Tay
- Department of Pathology, University of Utah, Salt Lake City, Utah
- Huntsman Cancer Institute, Salt Lake City, Utah
| | - Chris Stubben
- Bioinformatics Shared Resource, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Kenneth M. Boucher
- Cancer Biostatistics Shared Resource, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Juan M. Oviedo
- Department of Pathology, University of Utah, Salt Lake City, Utah
| | - Keke C. Fairfax
- Department of Pathology, University of Utah, Salt Lake City, Utah
| | - Matthew A. Williams
- Department of Pathology, University of Utah, Salt Lake City, Utah
- Huntsman Cancer Institute, Salt Lake City, Utah
| | - Sheri L. Holmen
- Huntsman Cancer Institute, Salt Lake City, Utah
- Department of Surgery, University of Utah, Salt Lake City, Utah
| | - Roger K. Wolff
- Department of Pathology, University of Utah, Salt Lake City, Utah
- Huntsman Cancer Institute, Salt Lake City, Utah
| | - Allie H. Grossmann
- Department of Pathology, University of Utah, Salt Lake City, Utah
- Huntsman Cancer Institute, Salt Lake City, Utah
- Lead contact
| |
Collapse
|
7
|
Ben Ahmed A, Lemaire Q, Scache J, Mariller C, Lefebvre T, Vercoutter-Edouart AS. O-GlcNAc Dynamics: The Sweet Side of Protein Trafficking Regulation in Mammalian Cells. Cells 2023; 12:1396. [PMID: 37408229 PMCID: PMC10216988 DOI: 10.3390/cells12101396] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 07/07/2023] Open
Abstract
The transport of proteins between the different cellular compartments and the cell surface is governed by the secretory pathway. Alternatively, unconventional secretion pathways have been described in mammalian cells, especially through multivesicular bodies and exosomes. These highly sophisticated biological processes rely on a wide variety of signaling and regulatory proteins that act sequentially and in a well-orchestrated manner to ensure the proper delivery of cargoes to their final destination. By modifying numerous proteins involved in the regulation of vesicular trafficking, post-translational modifications (PTMs) participate in the tight regulation of cargo transport in response to extracellular stimuli such as nutrient availability and stress. Among the PTMs, O-GlcNAcylation is the reversible addition of a single N-acetylglucosamine monosaccharide (GlcNAc) on serine or threonine residues of cytosolic, nuclear, and mitochondrial proteins. O-GlcNAc cycling is mediated by a single couple of enzymes: the O-GlcNAc transferase (OGT) which catalyzes the addition of O-GlcNAc onto proteins, and the O-GlcNAcase (OGA) which hydrolyses it. Here, we review the current knowledge on the emerging role of O-GlcNAc modification in the regulation of protein trafficking in mammalian cells, in classical and unconventional secretory pathways.
Collapse
|
8
|
Xu R, Wan M, Shi X, Ma S, Zhang L, Yi P, Zhang R. A Rab10-ACAP1-Arf6 GTPases cascade modulates M4 muscarinic acetylcholine receptor trafficking and signaling. Cell Mol Life Sci 2023; 80:87. [PMID: 36917255 PMCID: PMC11072986 DOI: 10.1007/s00018-023-04722-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 03/16/2023]
Abstract
Membrane trafficking processes regulate the G protein-coupled receptor activity. The muscarinic acetylcholine receptors (mAChRs) are highly pursued drug targets for neurological diseases, but the cellular machineries that control the trafficking of these receptors remain largely elusive. Here, we revealed the role of the small GTPase Rab10 as a negative regulator for the post-activation trafficking of M4 mAChR and the underlying mechanism. We show that constitutively active Rab10 arrests the receptor within Rab5-positive early endosomes and significantly hinders the resensitization of M4-mediated Ca2+ signaling. Mechanistically, M4 binds to Rab10-GTP, which requires the motif 386RKKRQMAA393 (R386-A393) within the third intracellular loop. Moreover, Rab10-GTP inactivates Arf6 by recruiting the Arf6 GTPase-activating protein, ACAP1. Strikingly, deletion of the motif R386-A393 causes M4 to bypass the control by Rab10 and switch to the Rab4-facilitated fast recycling pathway, thus reusing the receptor. Therefore, Rab10 couples the cargo sorting and membrane trafficking regulation through cycle between GTP-bound and GDP-bound state. Our findings suggest a model that Rab10 binds to the M4 like a molecular brake and controls the receptor's transport through endosomes, thus modulating the signaling, and this regulation is specific among the mAChR subtypes.
Collapse
Affiliation(s)
- Rongmei Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Min Wan
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, USA
| | - Xuemeng Shi
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
- College of Life Science, Henan Agricultural University, Zhengzhou, Henan, China
| | - Shumin Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lina Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ping Yi
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Rongying Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
9
|
Francis CR, Kincross H, Kushner EJ. Rab35 governs apicobasal polarity through regulation of actin dynamics during sprouting angiogenesis. Nat Commun 2022; 13:5276. [PMID: 36075898 PMCID: PMC9458672 DOI: 10.1038/s41467-022-32853-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 08/17/2022] [Indexed: 12/01/2022] Open
Abstract
In early blood vessel development, trafficking programs, such as those using Rab GTPases, are tasked with delivering vesicular cargo with high spatiotemporal accuracy. However, the function of many Rab trafficking proteins remain ill-defined in endothelial tissue; therefore, their relevance to blood vessel development is unknown. Rab35 has been shown to play an enigmatic role in cellular behaviors which differs greatly between tissue-type and organism. Importantly, Rab35 has never been characterized for its potential contribution in sprouting angiogenesis; thus, our goal was to map Rab35’s primary function in angiogenesis. Our results demonstrate that Rab35 is critical for sprout formation; in its absence, apicobasal polarity is entirely lost in vitro and in vivo. To determine mechanism, we systematically explored established Rab35 effectors and show that none are operative in endothelial cells. However, we find that Rab35 partners with DENNd1c, an evolutionarily divergent guanine exchange factor, to localize to actin. Here, Rab35 regulates actin polymerization through limiting Rac1 and RhoA activity, which is required to set up proper apicobasal polarity during sprout formation. Our findings establish that Rab35 is a potent brake of actin remodeling during blood vessel development. The promiscuous GTPase Rab35 has been shown to be involved in many important cellular functions. In this article, Francis et al. illustrate how Rab35 acts as a critical brake to actin remodeling during sprouting angiogenesis and how it is necessary for proper blood vessel development.
Collapse
Affiliation(s)
- Caitlin R Francis
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Hayle Kincross
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Erich J Kushner
- Department of Biological Sciences, University of Denver, Denver, CO, USA.
| |
Collapse
|
10
|
Hyper-Methylated Hub Genes of T-Cell Receptor Signaling Predict a Poor Clinical Outcome in Lung Adenocarcinoma. JOURNAL OF ONCOLOGY 2022; 2022:5426887. [PMID: 35432532 PMCID: PMC9007647 DOI: 10.1155/2022/5426887] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 11/17/2022]
Abstract
Background Immune checkpoint inhibitors (ICIs) emerge as the first-line treatment of lung adenocarcinoma (LUAD); selection of subpopulations acquiring clinical benefit is required. Associations between epigenetic modulation of tumor microenvironment (TME) and clinical outcome are far from clear. We focused on immune-related genes closely regulated by DNA methylation to identify the potential clinical outcome indicators. Methods We systematically calculated immunophenotype score (IMpS) and classified immunophenotypes based on seven TME features in three independent cohorts. The overlapping of differential expressed genes and methylated probes targeted genes was regarded as genes closely regulated by DNA methylation. Then, probe/gene pairs which highly correlated with each other and IMpS were identified and named as immune-related probe/gene pairs (mIMg). Prognostic mIMg were selected and verified in seven independent validation cohorts. Results Three immune phenotypes were clustered, and similar results were obtained in the three independent training cohorts. C2 displayed as an immunologically hot phenotype, whereas C3 corresponded with immunologically cold phenotype. Average methylation level was decreased from C2 to C3 (C2 > C1 > C3). Similarly, ICIs nonresponders showed global hypo-methylation compared with responders. Genes in mIMg were mainly enriched, especially in T-cell receptor activation, and repressed in noninflamed TME by hyper-methylation. Among mIMg, low expression and hyper-methylation of CD247, LCK, and PSTPIP1 were risk factors of overall survival (OS). ICIs nonresponders were more likely to be hyper-methylated in the three genes. By integrating with the oncogenes status, we demonstrated that EGFR wt and SRGN overexpressed patients were associated with chronic inflammation and immune evasion, showing an immunologically hot phenotype, which might lead to the short OS but derive clinical benefit from ICIs. Conclusions This study identifies hyper-methylation and concurrent repression of CD247, LCK, PSTPIP1 as immune negative indicators and risk factors for prognosis in LUAD. Moreover, EGFR/SRGN axis may participate in immune modification to influence ICIs response and clinical outcome in LUAD.
Collapse
|
11
|
Borghesan E, Smith EP, Myeni S, Binder K, Knodler LA, Celli J. A Brucella effector modulates the Arf6-Rab8a GTPase cascade to promote intravacuolar replication. EMBO J 2021; 40:e107664. [PMID: 34423453 PMCID: PMC8488576 DOI: 10.15252/embj.2021107664] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 01/15/2023] Open
Abstract
Remodeling of host cellular membrane transport pathways is a common pathogenic trait of many intracellular microbes that is essential to their intravacuolar life cycle and proliferation. The bacterium Brucella abortus generates a host endoplasmic reticulum‐derived vacuole (rBCV) that supports its intracellular growth, via VirB Type IV secretion system‐mediated delivery of effector proteins, whose functions and mode of action are mostly unknown. Here, we show that the effector BspF specifically promotes Brucella replication within rBCVs by interfering with vesicular transport between the trans‐Golgi network (TGN) and recycling endocytic compartment. BspF targeted the recycling endosome, inhibited retrograde traffic to the TGN, and interacted with the Arf6 GTPase‐activating Protein (GAP) ACAP1 to dysregulate Arf6‐/Rab8a‐dependent transport within the recycling endosome, which resulted in accretion of TGN‐associated vesicles by rBCVs and enhanced bacterial growth. Altogether, these findings provide mechanistic insight into bacterial modulation of membrane transport used to promote their own proliferation within intracellular vacuoles.
Collapse
Affiliation(s)
- Elizabeth Borghesan
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Erin P Smith
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Sebenzile Myeni
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Kelsey Binder
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Leigh A Knodler
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Jean Celli
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA.,Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| |
Collapse
|
12
|
In vitro reconstitution reveals phosphoinositides as cargo-release factors and activators of the ARF6 GAP ADAP1. Proc Natl Acad Sci U S A 2021; 118:2010054118. [PMID: 33443153 PMCID: PMC7817218 DOI: 10.1073/pnas.2010054118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The differentiation of cells depends on a precise control of their internal organization, which is the result of a complex dynamic interplay between the cytoskeleton, molecular motors, signaling molecules, and membranes. For example, in the developing neuron, the protein ADAP1 (ADP-ribosylation factor GTPase-activating protein [ArfGAP] with dual pleckstrin homology [PH] domains 1) has been suggested to control dendrite branching by regulating the small GTPase ARF6. Together with the motor protein KIF13B, ADAP1 is also thought to mediate delivery of the second messenger phosphatidylinositol (3,4,5)-trisphosphate (PIP3) to the axon tip, thus contributing to PIP3 polarity. However, what defines the function of ADAP1 and how its different roles are coordinated are still not clear. Here, we studied ADAP1's functions using in vitro reconstitutions. We found that KIF13B transports ADAP1 along microtubules, but that PIP3 as well as PI(3,4)P2 act as stop signals for this transport instead of being transported. We also demonstrate that these phosphoinositides activate ADAP1's enzymatic activity to catalyze GTP hydrolysis by ARF6. Together, our results support a model for the cellular function of ADAP1, where KIF13B transports ADAP1 until it encounters high PIP3/PI(3,4)P2 concentrations in the plasma membrane. Here, ADAP1 disassociates from the motor to inactivate ARF6, promoting dendrite branching.
Collapse
|
13
|
Jin Y, Meng Q, Zhang B, Xie C, Chen X, Tian B, Wang J, Shih TC, Zhang Y, Cao J, Yang Y, Chen S, Guan X, Chen X, Hong A. Cancer-associated fibroblasts-derived exosomal miR-3656 promotes the development and progression of esophageal squamous cell carcinoma via the ACAP2/PI3K-AKT signaling pathway. Int J Biol Sci 2021; 17:3689-3701. [PMID: 34671193 PMCID: PMC8495391 DOI: 10.7150/ijbs.62571] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/05/2021] [Indexed: 12/19/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common gastrointestinal tumors, accounting for almost half a million deaths per year. Cancer-associated fibroblasts (CAFs) are the major constituent of the tumor microenvironment (TME) and dramatically impact ESCC progression. Recent evidence suggests that exosomes derived from CAFs are able to transmit regulating signals and promote ESCC development. In this study, we compared different the component ratios of miRNAs in exosomes secreted by CAFs in tumors and with those from normal fibroblasts (NFs) in precancerous tissues. The mRNA level of hsa-miR-3656 was significantly upregulated in the former exosomes. Subsequently, by comparing tumor cell development in vitro and in vivo, we found that the proliferation, migration and invasion capabilities of ESCC cells were significantly improved when miR-3656 was present. Further target gene analysis confirmed ACAP2 was a target gene regulated by miR-3656 and exhibited a negative regulatory effect on tumor proliferation. Additionally, the downregulation of ACAP2 triggered by exosomal-derived miR-3656 further promotes the activation of the PI3K/AKT and β-catenin signaling pathways and ultimately improves the growth of ESCC cells both in vitro and in xenograft models. These results may represent a potential therapeutic target for ESCC and provide a new basis for clinical treatment plans.
Collapse
Affiliation(s)
- Yuan Jin
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, P. R. China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, P. R. China
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, P. R. China
- Guangdong Provincial biotechnology drug & Engineering Technology Research Center, Guangzhou 510632, P. R. China
| | - Qilin Meng
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, P. R. China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, P. R. China
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, P. R. China
- Guangdong Provincial biotechnology drug & Engineering Technology Research Center, Guangzhou 510632, P. R. China
| | - Bihui Zhang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, P. R. China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, P. R. China
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, P. R. China
- Guangdong Provincial biotechnology drug & Engineering Technology Research Center, Guangzhou 510632, P. R. China
| | - Chen Xie
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, P. R. China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, P. R. China
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, P. R. China
- Guangdong Provincial biotechnology drug & Engineering Technology Research Center, Guangzhou 510632, P. R. China
| | - Xue Chen
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, P. R. China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, P. R. China
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, P. R. China
- Guangdong Provincial biotechnology drug & Engineering Technology Research Center, Guangzhou 510632, P. R. China
| | - Baoqing Tian
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, P. R. China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, P. R. China
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, P. R. China
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, P. R. China
| | - Jiakang Wang
- Cancer Center of Guangzhou Medical University, Guangzhou 510090, P. R. China
| | - Tsung-Chieh Shih
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, California, USA
| | - Yibo Zhang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, P. R. China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, P. R. China
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, P. R. China
- Guangdong Provincial biotechnology drug & Engineering Technology Research Center, Guangzhou 510632, P. R. China
| | - Jieqiong Cao
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, P. R. China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, P. R. China
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, P. R. China
- Guangdong Provincial biotechnology drug & Engineering Technology Research Center, Guangzhou 510632, P. R. China
| | - Yiqi Yang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, P. R. China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, P. R. China
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, P. R. China
- Guangdong Provincial biotechnology drug & Engineering Technology Research Center, Guangzhou 510632, P. R. China
| | - Size Chen
- Oncology Department, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, P. R. China
- Guangdong Provincial Engineering Research Center for Precise Therapy of Esophageal Cancer, Guangzhou 510080, P. R. China
| | - Xinyuan Guan
- Department of Clinical Oncology, University of Hong Kong, Hong Kong, P. R. China
| | - Xiaojia Chen
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, P. R. China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, P. R. China
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, P. R. China
- Guangdong Provincial biotechnology drug & Engineering Technology Research Center, Guangzhou 510632, P. R. China
- ✉ Corresponding author: Dr. An Hong and Dr. Xiaojia Chen. (AH) , (XC)
| | - An Hong
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, P. R. China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, P. R. China
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, P. R. China
- Guangdong Provincial biotechnology drug & Engineering Technology Research Center, Guangzhou 510632, P. R. China
- ✉ Corresponding author: Dr. An Hong and Dr. Xiaojia Chen. (AH) , (XC)
| |
Collapse
|
14
|
Bhat S, Ljubojevic N, Zhu S, Fukuda M, Echard A, Zurzolo C. Rab35 and its effectors promote formation of tunneling nanotubes in neuronal cells. Sci Rep 2020; 10:16803. [PMID: 33033331 PMCID: PMC7544914 DOI: 10.1038/s41598-020-74013-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/10/2020] [Indexed: 12/11/2022] Open
Abstract
Tunneling nanotubes (TNTs) are F-actin rich structures that connect distant cells, allowing the transport of many cellular components, including vesicles, organelles and molecules. Rab GTPases are the major regulators of vesicle trafficking and also participate in actin cytoskeleton remodelling, therefore, we examined their role in TNTs. Rab35 functions with several proteins that are involved in vesicle trafficking such as ACAP2, MICAL-L1, ARF6 and EHD1, which are known to be involved in neurite outgrowth. Here we show that Rab35 promotes TNT formation and TNT-mediated vesicle transfer in a neuronal cell line. Furthermore, our data indicates that Rab35-GTP, ACAP2, ARF6-GDP and EHD1 act in a cascade mechanism to promote TNT formation. Interestingly, MICAL-L1 overexpression, shown to be necessary for the action of Rab35 on neurite outgrowth, showed no effect on TNTs, indicating that TNT formation and neurite outgrowth may be processed through similar but not identical pathways, further supporting the unique identity of these cellular protrusions.
Collapse
Affiliation(s)
- Shaarvari Bhat
- Unit of Membrane Traffic and Pathogenesis, UMR3691 CNRS, Institut Pasteur, 28 rue du Dr Roux, 75015, Paris, France
- Université Paris-Sud, Université Paris-Saclay, 91405, Orsay, France
| | - Nina Ljubojevic
- Unit of Membrane Traffic and Pathogenesis, UMR3691 CNRS, Institut Pasteur, 28 rue du Dr Roux, 75015, Paris, France
- Sorbonne Université, ED394-Physiologie, Physiopathologie et Thérapeutique, 75005, Paris, France
| | - Seng Zhu
- Unit of Membrane Traffic and Pathogenesis, UMR3691 CNRS, Institut Pasteur, 28 rue du Dr Roux, 75015, Paris, France
| | - Mitsunori Fukuda
- Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Arnaud Echard
- Membrane Traffic and Cell Division Lab, UMR3691 CNRS, Institut Pasteur, 75015, Paris, France
| | - Chiara Zurzolo
- Unit of Membrane Traffic and Pathogenesis, UMR3691 CNRS, Institut Pasteur, 28 rue du Dr Roux, 75015, Paris, France.
| |
Collapse
|
15
|
New Era of Diacylglycerol Kinase, Phosphatidic Acid and Phosphatidic Acid-Binding Protein. Int J Mol Sci 2020; 21:ijms21186794. [PMID: 32947951 PMCID: PMC7555651 DOI: 10.3390/ijms21186794] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022] Open
Abstract
Diacylglycerol kinase (DGK) phosphorylates diacylglycerol (DG) to generate phosphatidic acid (PA). Mammalian DGK consists of ten isozymes (α–κ) and governs a wide range of physiological and pathological events, including immune responses, neuronal networking, bipolar disorder, obsessive-compulsive disorder, fragile X syndrome, cancer, and type 2 diabetes. DG and PA comprise diverse molecular species that have different acyl chains at the sn-1 and sn-2 positions. Because the DGK activity is essential for phosphatidylinositol turnover, which exclusively produces 1-stearoyl-2-arachidonoyl-DG, it has been generally thought that all DGK isozymes utilize the DG species derived from the turnover. However, it was recently revealed that DGK isozymes, except for DGKε, phosphorylate diverse DG species, which are not derived from phosphatidylinositol turnover. In addition, various PA-binding proteins (PABPs), which have different selectivities for PA species, were recently found. These results suggest that DGK–PA–PABP axes can potentially construct a large and complex signaling network and play physiologically and pathologically important roles in addition to DGK-dependent attenuation of DG–DG-binding protein axes. For example, 1-stearoyl-2-docosahexaenoyl-PA produced by DGKδ interacts with and activates Praja-1, the E3 ubiquitin ligase acting on the serotonin transporter, which is a target of drugs for obsessive-compulsive and major depressive disorders, in the brain. This article reviews recent research progress on PA species produced by DGK isozymes, the selective binding of PABPs to PA species and a phosphatidylinositol turnover-independent DG supply pathway.
Collapse
|
16
|
Li Y, Lu S, Lan M, Peng X, Zhang Z, Lang J. A prognostic nomogram integrating novel biomarkers identified by machine learning for cervical squamous cell carcinoma. J Transl Med 2020; 18:223. [PMID: 32503630 PMCID: PMC7275455 DOI: 10.1186/s12967-020-02387-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023] Open
Abstract
Background Cervical cancer (CC) represents the fourth most frequently diagnosed malignancy affecting women all over the world. However, effective prognostic biomarkers are still limited for accurately identifying high-risk patients. Here, we provided a combination machine learning algorithm-based signature to predict the prognosis of cervical squamous cell carcinoma (CSCC). Methods and materials After utilizing RNA sequencing (RNA-seq) data from 36 formalin-fixed and paraffin-embedded (FFPE) samples, the most significant modules were highlighted by the weighted gene co-expression network analysis (WGCNA). A candidate genes-based prognostic classifier was constructed by the least absolute shrinkage and selection operator (LASSO) and then validated in an independent validation set. Finally, based on the multivariate analysis, a nomogram including the FIGO stage, therapy outcome, and risk score level was built to predict progression-free survival (PFS) probability. Results A mRNA-based signature was developed to classify patients into high- and low-risk groups with significantly different PFS and overall survival (OS) rate (training set: p < 0.001 for PFS, p = 0.016 for OS; validation set: p = 0.002 for PFS, p = 0.028 for OS). The prognostic classifier was an independent and powerful prognostic biomarker for PFS in both cohorts (training set: hazard ratio [HR] = 0.13, 95% CI 0.05–0.33, p < 0.001; validation set: HR = 0.02, 95% CI 0.01–0.04, p < 0.001). A nomogram that integrated the independent prognostic factors was constructed for clinical application. The calibration curve showed that the nomogram was able to predict 1-, 3-, and 5-year PFS accurately, and it performed well in the external validation cohorts (concordance index: 0.828 and 0.864, respectively). Conclusion The mRNA-based biomarker is a powerful and independent prognostic factor. Furthermore, the nomogram comprising our prognostic classifier is a promising predictor in identifying the progression risk of CSCC patients.
Collapse
Affiliation(s)
- Yimin Li
- School of Medicine, University of Electronic Science and Technology of China, No. 2006, Xiyuan Avenue, High-tech Zone (West District), Chengdu, 611731, Sichuan, People's Republic of China
| | - Shun Lu
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No. 55, South Renmin Avenue Fourth Section, Chengdu, 610041, Sichuan, People's Republic of China.,Radiation Oncology Key Laboratory of Sichuan Province, No. 55, South Renmin Avenue Fourth Section, Chengdu, 610041, Sichuan, People's Republic of China
| | - Mei Lan
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No. 55, South Renmin Avenue Fourth Section, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xinhao Peng
- School of Medicine, University of Electronic Science and Technology of China, No. 2006, Xiyuan Avenue, High-tech Zone (West District), Chengdu, 611731, Sichuan, People's Republic of China
| | - Zijian Zhang
- Department of Oncology, Xiangya Hospital Central South University, Kaifu District, Changsha, 410008, Hunan, People's Republic of China
| | - Jinyi Lang
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No. 55, South Renmin Avenue Fourth Section, Chengdu, 610041, Sichuan, People's Republic of China. .,Radiation Oncology Key Laboratory of Sichuan Province, No. 55, South Renmin Avenue Fourth Section, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
17
|
Zhang J, Zhang Q, Zhang J, Wang Q. Expression of ACAP1 Is Associated with Tumor Immune Infiltration and Clinical Outcome of Ovarian Cancer. DNA Cell Biol 2020; 39:1545-1557. [PMID: 32456571 DOI: 10.1089/dna.2020.5596] [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] [Indexed: 12/24/2022] Open
Abstract
ADP-ribosylation factor (Arf) GTPase-activating protein (GAP) with coiled-coil, ankyrin repeat and PH domains 1 (ACAP1) is an Arf6 GAP that regulates membrane trafficking and is critical for the migratory potential of cells. However, the roles of ACAP1 have not been fully explored and its association with clinicopathological features in ovarian cancer is still unknown. In this study, we systematically analyzed multiple databases, including TISIDB, Tumor Immune Estimation Resource (TIMER2.0), Gene Expression Omnibus (GEO), CORTECON, Kaplan-Meier Plotter and LinkedOmics platforms to reveal the clinical significance and function of ACAP1 in ovarian cancer. We found that the expression of ACAP1 was upregulated in ovarian cancer and high ACAP1 expression predicted poor prognosis. Our data demonstrated that the expression and methylation status of ACAP1 were strongly correlated with immune infiltration levels, immunomodulators, and chemokines. Gene set enrichment analysis (GSEA) analysis also showed that the mechanism of ACAP1 in regulating ovarian cancer was related to a variety of immune-related pathways. In addition, we also revealed that the expression of ACAP1 was altered during cell differentiation and associated with cancer cell stemness markers. Our study highlights the clinical significance of ACAP1 in ovarian cancer and provides insight into the novel function of ACAP1 in regulation of tumor immune microenvironment and cancer cell stemness.
Collapse
Affiliation(s)
- Jiawen Zhang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qinyi Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Jing Zhang
- Department of Integrated Therapy, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qingying Wang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
18
|
Mingirulli N, Pyle A, Hathazi D, Alston CL, Kohlschmidt N, O'Grady G, Waddell L, Evesson F, Cooper SBT, Turner C, Duff J, Topf A, Yubero D, Jou C, Nascimento A, Ortez C, García‐Cazorla A, Gross C, O'Callaghan M, Santra S, Preece MA, Champion M, Korenev S, Chronopoulou E, Anirban M, Pierre G, McArthur D, Thompson K, Navas P, Ribes A, Tort F, Schlüter A, Pujol A, Montero R, Sarquella G, Lochmüller H, Jiménez‐Mallebrera C, Taylor RW, Artuch R, Kirschner J, Grünert SC, Roos A, Horvath R. Clinical presentation and proteomic signature of patients with TANGO2 mutations. J Inherit Metab Dis 2020; 43:297-308. [PMID: 31339582 PMCID: PMC7078914 DOI: 10.1002/jimd.12156] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/17/2019] [Accepted: 07/18/2019] [Indexed: 12/20/2022]
Abstract
Transport And Golgi Organization protein 2 (TANGO2) deficiency has recently been identified as a rare metabolic disorder with a distinct clinical and biochemical phenotype of recurrent metabolic crises, hypoglycemia, lactic acidosis, rhabdomyolysis, arrhythmias, and encephalopathy with cognitive decline. We report nine subjects from seven independent families, and we studied muscle histology, respiratory chain enzyme activities in skeletal muscle and proteomic signature of fibroblasts. All nine subjects carried autosomal recessive TANGO2 mutations. Two carried the reported deletion of exons 3 to 9, one homozygous, one heterozygous with a 22q11.21 microdeletion inherited in trans. The other subjects carried three novel homozygous (c.262C>T/p.Arg88*; c.220A>C/p.Thr74Pro; c.380+1G>A), and two further novel heterozygous (c.6_9del/p.Phe6del); c.11-13delTCT/p.Phe5del mutations. Immunoblot analysis detected a significant decrease of TANGO2 protein. Muscle histology showed mild variation of fiber diameter, no ragged-red/cytochrome c oxidase-negative fibers and a defect of multiple respiratory chain enzymes and coenzyme Q10 (CoQ10 ) in two cases, suggesting a possible secondary defect of oxidative phosphorylation. Proteomic analysis in fibroblasts revealed significant changes in components of the mitochondrial fatty acid oxidation, plasma membrane, endoplasmic reticulum-Golgi network and secretory pathways. Clinical presentation of TANGO2 mutations is homogeneous and clinically recognizable. The hemizygous mutations in two patients suggest that some mutations leading to allele loss are difficult to detect. A combined defect of the respiratory chain enzymes and CoQ10 with altered levels of several membrane proteins provides molecular insights into the underlying pathophysiology and may guide rational new therapeutic interventions.
Collapse
Affiliation(s)
- Nadja Mingirulli
- Department of Neuropediatrics and Muscle DisordersMedical Center – University of Freiburg, Faculty of MedicineBreisgauGermany
- Department of General PediatricsAdolescent Medicine and Neonatology, Medical Center – University of Freiburg, Faculty of MedicineBreisgauGermany
| | - Angela Pyle
- Wellcome Centre for Mitochondrial ResearchInstitute of Genetic Medicine, Newcastle UniversityNewcastle upon TyneUK
| | - Denisa Hathazi
- Biomedical Research DepartmentLeibniz‐Institut für Analytische Wissenschaften – ISAS – e.VDortmundGermany
| | - Charlotte L. Alston
- Wellcome Centre for Mitochondrial ResearchInstitute of Neuroscience, Newcastle UniversityNewcastle upon TyneUK
| | | | - Gina O'Grady
- Kid's Neuroscience Centre, Children's Hospital at WestmeadSydneyNew South WalesAustralia
| | - Leigh Waddell
- Kid's Neuroscience Centre, Children's Hospital at WestmeadSydneyNew South WalesAustralia
| | - Frances Evesson
- Kid's Neuroscience Centre, Children's Hospital at WestmeadSydneyNew South WalesAustralia
- Discipline of Child and Adolescent HealthThe University of SydneySydneyNew South WalesAustralia
| | - Sandra B. T. Cooper
- Kid's Neuroscience Centre, Children's Hospital at WestmeadSydneyNew South WalesAustralia
- Discipline of Child and Adolescent HealthThe University of SydneySydneyNew South WalesAustralia
| | - Christian Turner
- Discipline of Child and Adolescent HealthThe University of SydneySydneyNew South WalesAustralia
- CardiologyThe Children's Hospital at WestmeadSydneyNew South WalesAustralia
| | - Jennifer Duff
- Wellcome Centre for Mitochondrial ResearchInstitute of Genetic Medicine, Newcastle UniversityNewcastle upon TyneUK
| | - Ana Topf
- John Walton Muscular Dystrophy Research CentreInstitute of Genetic Medicine, Newcastle UniversityNewcastle upon TyneUK
| | - Delia Yubero
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Cristina Jou
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Andrés Nascimento
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Carlos Ortez
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Angels García‐Cazorla
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Claudia Gross
- Wellcome Centre for Mitochondrial ResearchInstitute of Neuroscience, Newcastle UniversityNewcastle upon TyneUK
| | - Maria O'Callaghan
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Saikat Santra
- Birmingham Women's and Children's NHS Foundation TrustBirminghamUK
| | | | | | - Sergei Korenev
- Department of Inherited DiseaseSt Thomas HospitalLondonUK
| | | | - Majumdar Anirban
- South West Regional Metabolic DepartmentBristol Royal Hospital for ChildrenBristolUK
| | - Germaine Pierre
- South West Regional Metabolic DepartmentBristol Royal Hospital for ChildrenBristolUK
| | - Daniel McArthur
- Center for Mendelian Genomics and Program in Medical and Population GeneticsBroad Institute of MIT and HarvardCambridgeMassachusetts
- Analytic and Translational Genetics UnitMassachusetts General HospitalBostonMassachusetts
| | - Kyle Thompson
- Kid's Neuroscience Centre, Children's Hospital at WestmeadSydneyNew South WalesAustralia
| | - Placido Navas
- Centro Andaluz de Biología del DesarrolloUníversidad Pablo de Olavide‐CSIC‐JA and CIBERER, Instituto de Salud Carlos IIIMadridSpain
| | - Antonia Ribes
- Secció d'Errors Congènits del Metabolisme – IBCServei de Bioquímica I Genètìca Molecular, Hospital Clínìc, IDIBAPS, CIBERERBarcelonaSpain
| | - Frederic Tort
- Secció d'Errors Congènits del Metabolisme – IBCServei de Bioquímica I Genètìca Molecular, Hospital Clínìc, IDIBAPS, CIBERERBarcelonaSpain
| | - Agatha Schlüter
- Neurometabolic Diseases Laboratory, Institut d'Investìgacío Biomedíca de Bellvitge (IDIBELL), and Centre for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos IIIMadridSpain
| | - Aurora Pujol
- Catalan Institution of Research and Advanced Studies (ICREA)BarcelonaSpain
| | - Raquel Montero
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Georgia Sarquella
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Hanns Lochmüller
- Department of Neuropediatrics and Muscle DisordersMedical Center – University of Freiburg, Faculty of MedicineBreisgauGermany
- Children's Hospital of Eastern Ontario Research Institute, University of OttawaOttawaOntarioCanada
- Division of Neurology, Department of MedicineThe Ottawa HospitalOttawaOntarioCanada
| | - Cecilia Jiménez‐Mallebrera
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Robert W. Taylor
- Kid's Neuroscience Centre, Children's Hospital at WestmeadSydneyNew South WalesAustralia
| | - Rafael Artuch
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Janbernd Kirschner
- Department of Neuropediatrics and Muscle DisordersMedical Center – University of Freiburg, Faculty of MedicineBreisgauGermany
| | - Sarah C. Grünert
- Department of General PediatricsAdolescent Medicine and Neonatology, Medical Center – University of Freiburg, Faculty of MedicineBreisgauGermany
| | - Andreas Roos
- Biomedical Research DepartmentLeibniz‐Institut für Analytische Wissenschaften – ISAS – e.VDortmundGermany
- Pediatric NeurologyUniversity Children's Hospital, University of Duisburg‐Essen, Faculty of MedicineEssenGermany
| | - Rita Horvath
- Wellcome Centre for Mitochondrial ResearchInstitute of Genetic Medicine, Newcastle UniversityNewcastle upon TyneUK
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| |
Collapse
|
19
|
Chan C, Pang X, Zhang Y, Niu T, Yang S, Zhao D, Li J, Lu L, Hsu VW, Zhou J, Sun F, Fan J. ACAP1 assembles into an unusual protein lattice for membrane deformation through multiple stages. PLoS Comput Biol 2019; 15:e1007081. [PMID: 31291238 PMCID: PMC6663034 DOI: 10.1371/journal.pcbi.1007081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/29/2019] [Accepted: 05/06/2019] [Indexed: 11/19/2022] Open
Abstract
Studies on the Bin-Amphiphysin-Rvs (BAR) domain have advanced a fundamental understanding of how proteins deform membrane. We previously showed that a BAR domain in tandem with a Pleckstrin Homology (PH domain) underlies the assembly of ACAP1 (Arfgap with Coil-coil, Ankryin repeat, and PH domain I) into an unusual lattice structure that also uncovers a new paradigm for how a BAR protein deforms membrane. Here, we initially pursued computation-based refinement of the ACAP1 lattice to identify its critical protein contacts. Simulation studies then revealed how ACAP1, which dimerizes into a symmetrical structure in solution, is recruited asymmetrically to the membrane through dynamic behavior. We also pursued electron microscopy (EM)-based structural studies, which shed further insight into the dynamic nature of the ACAP1 lattice assembly. As ACAP1 is an unconventional BAR protein, our findings broaden the understanding of the mechanistic spectrum by which proteins assemble into higher-ordered structures to achieve membrane deformation.
Collapse
Affiliation(s)
- Chun Chan
- Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong, China
| | - Xiaoyun Pang
- National Laboratory of Biomacromolecules, CAS Center for excellence in biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yan Zhang
- National Laboratory of Biomacromolecules, CAS Center for excellence in biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Tongxin Niu
- National Laboratory of Biomacromolecules, CAS Center for excellence in biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Shengjiang Yang
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Daohui Zhao
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Jian Li
- Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, and Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lanyuan Lu
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Victor W. Hsu
- Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, and Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jian Zhou
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, Guangdong, China
- * E-mail: (JZ); (FS); (JF)
| | - Fei Sun
- National Laboratory of Biomacromolecules, CAS Center for excellence in biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Center for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- * E-mail: (JZ); (FS); (JF)
| | - Jun Fan
- Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Center for Advanced Nuclear Safety and Sustainable Development, City University of Hong Kong, Hong Kong, China
- * E-mail: (JZ); (FS); (JF)
| |
Collapse
|
20
|
Oncogenic lncRNA downregulates cancer cell antigen presentation and intrinsic tumor suppression. Nat Immunol 2019; 20:835-851. [PMID: 31160797 PMCID: PMC6619502 DOI: 10.1038/s41590-019-0400-7] [Citation(s) in RCA: 252] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 04/12/2019] [Indexed: 12/12/2022]
Abstract
The mechanisms through which tumor cells genetically lose antigenicity and evade immune checkpoints remain largely elusive. Here, we report that tissue-specific expression of the human long-noncoding RNA LINK-A in mouse mammary glands initiated metastatic mammary gland tumors, which phenotypically resembled human triple-negative breast cancer (TNBC). LINK-A expression facilitated crosstalk between phosphatidylinositol-(3,4,5)-trisphosphate and inhibitory G-protein–coupled receptor (GPCR) pathways, attenuating protein kinase A (PKA)-mediated phosphorylation of the E3 ubiquitin ligase TRIM71. Consequently, LINK-A expression enhanced K48–polyubiquitination-mediated degradation of the antigen peptide-loading complex (PLC) and intrinsic tumor suppressors Rb and p53. Treatment with LINK-A-locked nucleic acids or GPCR antagonists stabilized the PLC components, Rb, and p53, and sensitized mammary gland tumors to immune checkpoint blockers (ICBs). Importantly, PD-1 blockade-resistant TNBC patients exhibited elevated LINK-A levels and downregulated PLC components. Hence, we demonstrated lncRNA-dependent downregulation of antigenicity and intrinsic tumor suppression, which may provide the basis for developing a therapeutic regimen of combinational immunotherapy and effective early prevention for TNBCs.
Collapse
|
21
|
Yoo JH, Brady SW, Acosta-Alvarez L, Rogers A, Peng J, Sorensen LK, Wolff RK, Mleynek T, Shin D, Rich CP, Kircher DA, Bild A, Odelberg SJ, Li DY, Holmen SL, Grossmann AH. The Small GTPase ARF6 Activates PI3K in Melanoma to Induce a Prometastatic State. Cancer Res 2019; 79:2892-2908. [PMID: 31048499 DOI: 10.1158/0008-5472.can-18-3026] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 01/11/2019] [Accepted: 04/09/2019] [Indexed: 12/17/2022]
Abstract
Melanoma has an unusual capacity to spread in early-stage disease, prompting aggressive clinical intervention in very thin primary tumors. Despite these proactive efforts, patients with low-risk, low-stage disease can still develop metastasis, indicating the presence of permissive cues for distant spread. Here, we show that constitutive activation of the small GTPase ARF6 (ARF6Q67L) is sufficient to accelerate metastasis in mice with BRAFV600E/Cdkn2aNULL melanoma at a similar incidence and severity to Pten loss, a major driver of PI3K activation and melanoma metastasis. ARF6Q67L promoted spontaneous metastasis from significantly smaller primary tumors than PTENNULL, implying an enhanced ability of ARF6-GTP to drive distant spread. ARF6 activation increased lung colonization from circulating melanoma cells, suggesting that the prometastatic function of ARF6 extends to late steps in metastasis. Unexpectedly, ARF6Q67L tumors showed upregulation of Pik3r1 expression, which encodes the p85 regulatory subunit of PI3K. Tumor cells expressing ARF6Q67L displayed increased PI3K protein levels and activity, enhanced PI3K distribution to cellular protrusions, and increased AKT activation in invadopodia. ARF6 is necessary and sufficient for activation of both PI3K and AKT, and PI3K and AKT are necessary for ARF6-mediated invasion. We provide evidence for aberrant ARF6 activation in human melanoma samples, which is associated with reduced survival. Our work reveals a previously unknown ARF6-PI3K-AKT proinvasive pathway, it demonstrates a critical role for ARF6 in multiple steps of the metastatic cascade, and it illuminates how melanoma cells can acquire an early metastatic phenotype in patients. SIGNIFICANCE: These findings reveal a prometastatic role for ARF6 independent of tumor growth, which may help explain how melanoma spreads distantly from thin, early-stage primary tumors.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/11/2892/F1.large.jpg.
Collapse
Affiliation(s)
- Jae Hyuk Yoo
- Department of Medicine, Program in Molecular Medicine, University of Utah, Salt Lake City, Utah
| | - Samuel W Brady
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, Utah.,Department of Biomedical Informatics, School of Medicine, University of Utah, Salt Lake City, Utah
| | | | - Aaron Rogers
- Department of Pathology, University of Utah, Salt Lake City, Utah
| | - Jingfu Peng
- Department of Pathology, University of Utah, Salt Lake City, Utah
| | - Lise K Sorensen
- Department of Medicine, Program in Molecular Medicine, University of Utah, Salt Lake City, Utah
| | - Roger K Wolff
- Department of Pathology, University of Utah, Salt Lake City, Utah
| | - Tara Mleynek
- Department of Medicine, Program in Molecular Medicine, University of Utah, Salt Lake City, Utah
| | - Donghan Shin
- Department of Medicine, Program in Molecular Medicine, University of Utah, Salt Lake City, Utah
| | - Coulson P Rich
- Department of Pathology, University of Utah, Salt Lake City, Utah.,Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - David A Kircher
- Department of Oncological Sciences, School of Medicine, University of Utah, Salt Lake City, Utah
| | - Andrea Bild
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, Utah.,Department of Oncological Sciences, School of Medicine, University of Utah, Salt Lake City, Utah.,Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Institute, Monrovia, California
| | - Shannon J Odelberg
- Department of Medicine, Program in Molecular Medicine, University of Utah, Salt Lake City, Utah.,Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah.,Division of Cardiovascular Medicine, Department of Medicine, University of Utah, Salt Lake City, Utah
| | - Dean Y Li
- Department of Medicine, Program in Molecular Medicine, University of Utah, Salt Lake City, Utah.,Division of Cardiovascular Medicine, Department of Medicine, University of Utah, Salt Lake City, Utah.,Department of Human Genetics, University of Utah, Salt Lake City, Utah
| | - Sheri L Holmen
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah.,Department of Oncological Sciences, School of Medicine, University of Utah, Salt Lake City, Utah.,Department of Surgery, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Allie H Grossmann
- Department of Pathology, University of Utah, Salt Lake City, Utah. .,Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah.,ARUP Laboratories, University of Utah, Salt Lake City, Utah
| |
Collapse
|
22
|
Lin L, Shi Y, Wang M, Wang C, Zhu J, Zhang R. Rab35/ACAP2 and Rab35/RUSC2 Complex Structures Reveal Molecular Basis for Effector Recognition by Rab35 GTPase. Structure 2019; 27:729-740.e3. [PMID: 30905672 DOI: 10.1016/j.str.2019.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/03/2019] [Accepted: 02/24/2019] [Indexed: 11/16/2022]
Abstract
Rab35, a master regulator of membrane trafficking, regulates diverse cellular processes and is associated with various human diseases. Although a number of effectors have been identified, the molecular basis of Rab35-effector interactions remains unclear. Here, we provide the high-resolution crystal structures of Rab35 in complex with its two specific effectors ACAP2 and RUSC2, respectively. In the Rab35/ACAP2 complex structure, Rab35 binds to the terminal ankyrin repeat and a C-terminal extended α helix of ACAP2, revealing a previously uncharacterized binding mode both for Rabs and ankyrin repeats. In the Rab35/RUSC2 complex structure, Arg1015 of RUSC2 functions as a "pseudo-arginine finger" that stabilizes the GTP-bound Rab35, thus facilitating the assembly of Rab35/RUSC2 complex. The structural analysis allows us to design specific Rab35 mutants capable of eliminating Rab35/ACAP2 and Rab35/RUSC2 interactions, but not interfering with other effector bindings. The atomic structures also offer possible explanations to disease-associated mutants identified at the Rab35-effector interfaces.
Collapse
Affiliation(s)
- Lin Lin
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai Science Research Center, 333 Haike Road, Shanghai 201210, China
| | - Yingdong Shi
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Mengli Wang
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Chao Wang
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Jinwei Zhu
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai Science Research Center, 333 Haike Road, Shanghai 201210, China.
| | - Rongguang Zhang
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai Science Research Center, 333 Haike Road, Shanghai 201210, China.
| |
Collapse
|
23
|
Wang YC, Westcott NP, Griffin ME, Hang HC. Peptidoglycan Metabolite Photoaffinity Reporters Reveal Direct Binding to Intracellular Pattern Recognition Receptors and Arf GTPases. ACS Chem Biol 2019; 14:405-414. [PMID: 30735346 DOI: 10.1021/acschembio.8b01038] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The peptidoglycan fragments γ-d-glutamyl- meso-diaminopimelic acid (iE-DAP) and muramyl-dipeptide (MDP) are microbial-specific metabolites that activate intracellular pattern recognition receptors and stimulate immune signaling pathways. While extensive structure-activity studies have demonstrated that these bacterial cell wall metabolites trigger NOD1- and NOD2-dependent signaling, their direct binding to these innate immune receptors or other proteins in mammalian cells has not been established. To characterize these fundamental microbial metabolite-host interactions, we synthesized a series of peptidoglycan metabolite photoaffinity reporters and evaluated their cross-linking to NOD1 and NOD2 in mammalian cells. We show that active iE-DAP and MDP photoaffinity reporters selectively cross-linked NOD1 and NOD2, respectively, and not their inactive mutants. We also discovered MDP reporter cross-linking to Arf GTPases, which interacted most prominently with GTP-bound Arf6 and coimmunoprecipitated with NOD2 upon MDP stimulation. Notably, MDP binding to NOD2 and Arf6 was abrogated with loss-of-function NOD2 mutants associated with Crohn's disease. Our studies demonstrate peptidoglycan metabolite photoaffinity reporters can capture their cognate immune receptors in cells and reveal unpredicted ligand-induced interactions with other cellular cofactors. These photoaffinity reporters should afford useful tools to discover and characterize other peptidoglycan metabolite-interacting proteins.
Collapse
Affiliation(s)
- Yen-Chih Wang
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York, New York 10065, United States
| | - Nathan P. Westcott
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York, New York 10065, United States
| | - Matthew E. Griffin
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York, New York 10065, United States
| | - Howard C. Hang
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York, New York 10065, United States
| |
Collapse
|
24
|
Tanna CE, Goss LB, Ludwig CG, Chen PW. Arf GAPs as Regulators of the Actin Cytoskeleton-An Update. Int J Mol Sci 2019; 20:ijms20020442. [PMID: 30669557 PMCID: PMC6358971 DOI: 10.3390/ijms20020442] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 12/25/2022] Open
Abstract
Arf GTPase-activating proteins (Arf GAPs) control the activity of ADP-ribosylation factors (Arfs) by inducing GTP hydrolysis and participate in a diverse array of cellular functions both through mechanisms that are dependent on and independent of their Arf GAP activity. A number of these functions hinge on the remodeling of actin filaments. Accordingly, some of the effects exerted by Arf GAPs involve proteins known to engage in regulation of the actin dynamics and architecture, such as Rho family proteins and nonmuscle myosin 2. Circular dorsal ruffles (CDRs), podosomes, invadopodia, lamellipodia, stress fibers and focal adhesions are among the actin-based structures regulated by Arf GAPs. Arf GAPs are thus important actors in broad functions like adhesion and motility, as well as the specialized functions of bone resorption, neurite outgrowth, and pathogen internalization by immune cells. Arf GAPs, with their multiple protein-protein interactions, membrane-binding domains and sites for post-translational modification, are good candidates for linking the changes in actin to the membrane. The findings discussed depict a family of proteins with a critical role in regulating actin dynamics to enable proper cell function.
Collapse
Affiliation(s)
- Christine E Tanna
- Department of Biology, Williams College, Williamstown, MA 01267, USA.
| | - Louisa B Goss
- Department of Biology, Williams College, Williamstown, MA 01267, USA.
| | - Calvin G Ludwig
- Department of Biology, Williams College, Williamstown, MA 01267, USA.
| | - Pei-Wen Chen
- Department of Biology, Williams College, Williamstown, MA 01267, USA.
| |
Collapse
|
25
|
Zobel M, Disanza A, Senic-Matuglia F, Franco M, Colaluca IN, Confalonieri S, Bisi S, Barbieri E, Caldieri G, Sigismund S, Pece S, Chavrier P, Di Fiore PP, Scita G. A NUMB-EFA6B-ARF6 recycling route controls apically restricted cell protrusions and mesenchymal motility. J Cell Biol 2018; 217:3161-3182. [PMID: 30061108 PMCID: PMC6123001 DOI: 10.1083/jcb.201802023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/17/2018] [Accepted: 06/05/2018] [Indexed: 12/13/2022] Open
Abstract
The endocytic protein NUMB has been implicated in the control of various polarized cellular processes, including the acquisition of mesenchymal migratory traits through molecular mechanisms that have only been partially defined. Here, we report that NUMB is a negative regulator of a specialized set of understudied, apically restricted, actin-based protrusions, the circular dorsal ruffles (CDRs), induced by either PDGF or HGF stimulation. Through its PTB domain, NUMB binds directly to an N-terminal NPLF motif of the ARF6 guanine nucleotide exchange factor, EFA6B, and promotes its exchange activity in vitro. In cells, a NUMB-EFA6B-ARF6 axis regulates the recycling of the actin regulatory cargo RAC1 and is critical for the formation of CDRs that mark the acquisition of a mesenchymal mode of motility. Consistently, loss of NUMB promotes HGF-induced cell migration and invasion. Thus, NUMB negatively controls membrane protrusions and the acquisition of mesenchymal migratory traits by modulating EFA6B-ARF6 activity.
Collapse
Affiliation(s)
- Martina Zobel
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Andrea Disanza
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | | | - Michel Franco
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | | | | | - Sara Bisi
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Elisa Barbieri
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Giusi Caldieri
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Sara Sigismund
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Salvatore Pece
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Philippe Chavrier
- Institut Curie, PSL Research University, Paris, France
- Centre National de la Recherche Scientifique UMR 144, Membrane and Cytoskeleton Dynamics Team, Paris, France
| | - Pier Paolo Di Fiore
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giorgio Scita
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
26
|
Nieuwenhuis B, Haenzi B, Andrews MR, Verhaagen J, Fawcett JW. Integrins promote axonal regeneration after injury of the nervous system. Biol Rev Camb Philos Soc 2018; 93:1339-1362. [PMID: 29446228 PMCID: PMC6055631 DOI: 10.1111/brv.12398] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 12/23/2017] [Accepted: 01/11/2018] [Indexed: 12/13/2022]
Abstract
Integrins are cell surface receptors that form the link between extracellular matrix molecules of the cell environment and internal cell signalling and the cytoskeleton. They are involved in several processes, e.g. adhesion and migration during development and repair. This review focuses on the role of integrins in axonal regeneration. Integrins participate in spontaneous axonal regeneration in the peripheral nervous system through binding to various ligands that either inhibit or enhance their activation and signalling. Integrin biology is more complex in the central nervous system. Integrins receptors are transported into growing axons during development, but selective polarised transport of integrins limits the regenerative response in adult neurons. Manipulation of integrins and related molecules to control their activation state and localisation within axons is a promising route towards stimulating effective regeneration in the central nervous system.
Collapse
Affiliation(s)
- Bart Nieuwenhuis
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW)1105 BAAmsterdamThe Netherlands
| | - Barbara Haenzi
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
| | | | - Joost Verhaagen
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW)1105 BAAmsterdamThe Netherlands
- Centre for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVrije Universiteit Amsterdam1081 HVAmsterdamThe Netherlands
| | - James W. Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
- Centre of Reconstructive NeuroscienceInstitute of Experimental Medicine142 20Prague 4Czech Republic
| |
Collapse
|
27
|
Abstract
The maintenance of cell surface proteins is critical to the ability of a cell to sense and respond to information in its environment. As such, modulation of cell surface composition and receptor trafficking is a potentially important target of control in virus infection. Sorting endosomes (SEs) are control stations regulating the recycling or degradation of internalized plasma membrane proteins. Here we report that human cytomegalovirus (HCMV), a ubiquitous betaherpesvirus, alters the fate of internalized clathrin-independent endocytosis (CIE) cargo proteins, retaining them in virally reprogrammed SEs. We show that the small G protein ARF6 (ADP ribosylation factor 6), a regulator of CIE trafficking, is highly associated with SE membranes relative to uninfected cells. Combined with the observation of accumulated CIE cargo at the SE, these results suggest that infection diminishes the egress of ARF6 and its cargo from the SE. Expression of ubiquitin-specific protease 6 (USP6), also known as TRE17, was sufficient to restore ARF6 and some ARF6 cargo trafficking to the cell surface in infected cells. The USP activity of TRE17 was required to rescue both ARF6 and associated cargo from SE retention in infection. The finding that TRE17 expression does not rescue the trafficking of all CIE cargos retained at SEs in infection suggests that HCMV hijacks the normal sorting machinery and selectively sorts specific cargos into endocytic microdomains that are subject to alternative sorting fates. Cells maintain their surface composition, take up nutrients, and respond to their environment through the internalization and recycling of cargo at the cell surface through endocytic trafficking pathways. During infection with human cytomegalovirus (HCMV), host endocytic membranes are reorganized into a juxtanuclear structure associated with viral assembly and egress. Less appreciated is the effect of this reorganization on the trafficking of host proteins through the endocytic pathway. We show that HCMV retains internalized cargo and the effector of clathrin-independent endocytosis at sorting endosomes. The retention of some cargo, but not all, was reversed by overexpression of a ubiquitin-specific protease, TRE17. Our results demonstrate that HCMV induces profound reprogramming of endocytic trafficking and influences cargo sorting decisions. Further, our work suggests the presence of a novel ubiquitin-regulated checkpoint for the recycling of cargo from sorting endosome. These findings have important implications for host signaling and immune pathways in the context of HCMV infection.
Collapse
|
28
|
Naramoto S, Kyozuka J. ARF GTPase machinery at the plasma membrane regulates auxin transport-mediated plant growth. PLANT BIOTECHNOLOGY (TOKYO, JAPAN) 2018; 35:155-159. [PMID: 31819717 PMCID: PMC6879391 DOI: 10.5511/plantbiotechnology.18.0312a] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 03/12/2018] [Indexed: 05/23/2023]
Abstract
VAN3 is a plant ACAP-type ADP-ribosylation factor-GTPase activating protein (ARF-GAP) that regulates auxin transport-mediated plant morphogenesis such as continuous venation and lateral root development in Arabidopsis. Previous studies suggested that VAN3 localizes at the plasma membrane (PM) and intracellular structures. However, the role of PM localization in mediating the van3 mutant phenotype is not clear. Here we performed subcellular localization analysis of VAN3 and its regulators CVP2 and VAB to determine their endogenous functions. We found that GFP-tagged CVP2 and VAB preferentially localize at the PM in stably transformed plants. We determined that transgenic plants with lower expression levels of GFP- or mRFP-tagged VAN3 displayed PM localization, which was sufficient to rescue the van3 mutant. Functional VAN3-mRFP and VAB-GFP colocalized at PMs. The van3 mutant phenotype was suppressed by mutation of VAN7/GNOM, which encodes an ARF-GEF that localizes at the PM and Golgi apparatus. These combined results suggest that ARF-GTPase machinery at the PM regulates auxin transport-mediated plant growth and development.
Collapse
Affiliation(s)
- Satoshi Naramoto
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8577, Japan
| | - Junko Kyozuka
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8577, Japan
| |
Collapse
|
29
|
Neurotrophin Responsiveness of Sympathetic Neurons Is Regulated by Rapid Mobilization of the p75 Receptor to the Cell Surface through TrkA Activation of Arf6. J Neurosci 2018; 38:5606-5619. [PMID: 29789375 DOI: 10.1523/jneurosci.0788-16.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/24/2018] [Accepted: 05/13/2018] [Indexed: 12/23/2022] Open
Abstract
The p75 neurotrophin receptor (p75NTR) plays an integral role in patterning the sympathetic nervous system during development. Initially, p75NTR is expressed at low levels as sympathetic axons project toward their targets, which enables neurotrophin-3 (NT3) to activate TrkA receptors and promote growth. Upon reaching nerve growth factor (NGF) producing tissues, p75NTR is upregulated, resulting in formation of TrkA-p75 complexes, which are high-affinity binding sites selective for NGF, thereby blunting NT3 signaling. The level of p75NTR expressed on the neuron surface is instrumental in regulating trophic factor response; however, the mechanisms by which p75NTR expression is regulated are poorly understood. Here, we demonstrate a rapid, translation independent increase in surface expression of p75NTR in response to NGF in rat sympathetic neurons. p75NTR was mobilized to the neuron surface from GGA3-postitive vesicles through activation of the GTPase Arf6, which was stimulated by NGF, but not NT3 binding to TrkA. Arf6 activation required PI3 kinase activity and was prevented by an inhibitor of the cytohesin family of Arf6 guanine nucleotide exchange factors. Overexpression of a constitutively active Arf6 mutant (Q67L) was sufficient to significantly increase surface expression of p75NTR even in the absence of NGF. Functionally, expression of active Arf6 markedly attenuated the ability of NT3 to promote neuronal survival and neurite outgrowth, whereas the NGF response was unaltered. These data suggest that NGF activation of Arf6 through TrkA is critical for the increase in p75NTR surface expression that enables the switch in neurotrophin responsiveness during development in the sympathetic nervous system.SIGNIFICANCE STATEMENT p75NTR is instrumental in the regulation of neuronal survival and apoptosis during development and is also implicated as a contributor to aberrant neurodegeneration in numerous conditions. Therefore, a better understanding of the mechanisms that mediate p75NTR surface availability may provide insight into how and why neurodegenerative processes manifest and reveal new therapeutic targets. Results from this study indicate a novel mechanism by which p75NTR can be rapidly shuttled to the cell surface from existing intracellular pools and explores a unique pathway by which NGF regulates the sympathetic innervation of target tissues, which has profound consequences for the function of these organs.
Collapse
|
30
|
Chua CEL, Tang BL. Rab 10-a traffic controller in multiple cellular pathways and locations. J Cell Physiol 2018; 233:6483-6494. [PMID: 29377137 DOI: 10.1002/jcp.26503] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/24/2018] [Indexed: 12/29/2022]
Abstract
Rab GTPases are key regulators of eukaryotic membrane traffic, and their functions and activities are limited to particular intracellular transport steps and their membrane localization is by and large restricted. Some Rabs do participate in more than one transport steps, but broadly speaking, there is a clear demarcation between exocytic and endocytic Rabs. One Rab protein, Rab10, however, appears to be anomalous in this regard and has a diverse array of functions and subcellular localizations. Rab10 has been implicated in a myriad of activities ranging from polarized exocytosis and endosomal sorting in polarized cells, insulin-dependent Glut4 transport in adipocytes, axonal growth in neurons, and endo-phagocytic processes in macrophages. It's reported subcellular localizations include the endoplasmic reticulum (ER), Golgi/TGN, the endosomes/phagosomes and the primary cilia. In this review, we summarize and discuss the multitude of known roles of Rab10 in cellular membrane transport and the molecular players and mechanisms associated with these roles.
Collapse
Affiliation(s)
- Christelle En Lin Chua
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| | - Bor L Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| |
Collapse
|
31
|
Guo S, Fan XF, Jin JY, Fan LL, Zeng L, Zhou ZB, Xiang R, Tang JY. A novel proximal 3q29 chromosome microdeletion in a Chinese patient with Chiari malformation type II and Sprengel's deformity. Mol Cytogenet 2018; 11:8. [PMID: 29410707 PMCID: PMC5781338 DOI: 10.1186/s13039-018-0358-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/15/2018] [Indexed: 01/29/2023] Open
Abstract
Background Chiari malformation type II (CM-II) is mainly characterized by elongation and descent of the cerebellum through the foramen magnum into the spinal canal. Moreover, CM-II is uniquely associated with myelomeningocele. Sprengel’s deformity refers to the malposition of the scapula, i.e. scapular elevation which is sometimes accompanied with scapula dysplasia. Although few familial cases of CM-II and Sprengel’s deformity have been previously reported, both of these defects are considered to be sporadic, thus the exact etiology and causative genes have largely remained unknown. Case presentation The patient was diagnosed with CM-II accompanied with Sprengel’s deformity. Further genetic investigation revealed a novel 666 kb microdeletion located in 3q29 (chr3:194,532,035–195,198,585; Hg19). Subsequently, genes within the affected region were summarized, and XXYLT1 and ACAP2 were identified as the candidate genes. Conclusion We reported a case of a patient with CM-II and Sprengel’s deformity harboring a microdeletion in 3q29. This case highlights the importance of 3q29 in early neural and skeletal development, as well as expands the phenotype spectrum of this rare disorder.
Collapse
Affiliation(s)
- Shuai Guo
- 1Department of orthopaedics, Xiangya Hospital of Central South University, Changsha, 410078 People's Republic of China.,2School of Life Sciences, Central South University, Changsha, 410013 People's Republic of China
| | - Xue-Feng Fan
- 1Department of orthopaedics, Xiangya Hospital of Central South University, Changsha, 410078 People's Republic of China
| | - Jie-Yuan Jin
- 2School of Life Sciences, Central South University, Changsha, 410013 People's Republic of China
| | - Liang-Liang Fan
- 2School of Life Sciences, Central South University, Changsha, 410013 People's Republic of China
| | - Lei Zeng
- 1Department of orthopaedics, Xiangya Hospital of Central South University, Changsha, 410078 People's Republic of China
| | - Zheng-Bing Zhou
- 1Department of orthopaedics, Xiangya Hospital of Central South University, Changsha, 410078 People's Republic of China
| | - Rong Xiang
- 1Department of orthopaedics, Xiangya Hospital of Central South University, Changsha, 410078 People's Republic of China.,2School of Life Sciences, Central South University, Changsha, 410013 People's Republic of China
| | - Ju-Yu Tang
- 1Department of orthopaedics, Xiangya Hospital of Central South University, Changsha, 410078 People's Republic of China
| |
Collapse
|
32
|
Luo R, Reed CE, Sload JA, Wordeman L, Randazzo PA, Chen PW. Arf GAPs and molecular motors. Small GTPases 2017; 10:196-209. [PMID: 28430047 DOI: 10.1080/21541248.2017.1308850] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Arf GTPase-activating proteins (Arf GAPs) were first identified as regulators of the small GTP-binding proteins ADP-ribosylation factors (Arfs). The Arf GAPs are a large family of proteins in metazoans, outnumbering the Arfs that they regulate. The members of the Arf GAP family have complex domain structures and some have been implicated in particular cellular functions, such as cell migration, or with particular pathologies, such as tumor invasion and metastasis. The specific effects of Arfs sometimes depend on the Arf GAP involved in their regulation. These observations have led to speculation that the Arf GAPs themselves may affect cellular activities in capacities beyond the regulation of Arfs. Recently, 2 Arf GAPs, ASAP1 and AGAP1, have been found to bind directly to and influence the activity of myosins and kinesins, motor proteins associated with filamentous actin and microtubules, respectively. The Arf GAP-motor protein interaction is critical for cellular behaviors involving the actin cytoskeleton and microtubules, such as cell migration and other cell movements. Arfs, then, may function with molecular motors through Arf GAPs to regulate microtubule and actin remodeling.
Collapse
Affiliation(s)
- Ruibai Luo
- a Laboratory of Cellular and Molecular Biology , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Christine E Reed
- c Department of Biology , Williams College , Williamstown , MA , USA
| | - Jeffrey A Sload
- c Department of Biology , Williams College , Williamstown , MA , USA
| | - Linda Wordeman
- b Department of Physiology and Biophysics , University of Washington School of Medicine , Seattle , WA , USA
| | - Paul A Randazzo
- a Laboratory of Cellular and Molecular Biology , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Pei-Wen Chen
- c Department of Biology , Williams College , Williamstown , MA , USA
| |
Collapse
|
33
|
Chan KC, Lu L, Sun F, Fan J. Molecular Details of the PH Domain of ACAP1BAR-PH Protein Binding to PIP-Containing Membrane. J Phys Chem B 2017; 121:3586-3596. [DOI: 10.1021/acs.jpcb.6b09563] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Kevin Chun Chan
- Department
of Physics and Materials Science, City University of Hong Kong, Hong Kong, China
| | - Lanyuan Lu
- School
of Biological Sciences, Nanyang Technological University, 639798, Singapore
| | - Fei Sun
- National
Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Center
for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jun Fan
- Department
of Physics and Materials Science, City University of Hong Kong, Hong Kong, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
- Center for
Advanced Nuclear Safety and Sustainable Development, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
34
|
Yoo CM, Naramoto S, Sparks JA, Khan BR, Nakashima J, Fukuda H, Blancaflor EB. Deletion analysis of AGD1 reveals domains crucial for its plasma membrane recruitment and function in root hair polarity. J Cell Sci 2017. [DOI: 10.1242/jcs.203828] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
AGD1, a plant ACAP-type ADP-ribosylation factor-GTPase activating protein (ARF-GAP), functions in specifying root hair polarity in Arabidopsis thaliana. To better understand how AGD1 modulates root hair growth, we generated full length and domain-deleted AGD1-green fluorescent protein (GFP) constructs, and followed their localization during root hair development. AGD1-GFP localized to the cytoplasm and was recruited to specific regions of the root hair plasma membrane (PM). Distinct PM AGD1-GFP signal was first detected along the site of root hair bulge formation. The construct continued to mark the PM at the root hair apical dome but only during periods of reduced growth. During rapid tip-growth, AGD1-GFP labeled the PM of the lateral flanks and dissipated from the apical-most PM. Deletion analysis and a single domain GFP fusion revealed that the pleckstrin homology (PH) domain is the minimal unit required for recruitment of AGD1 to the PM. Our results indicate that differential recruitment of AGD1 to specific PM domains is an essential component of the membrane trafficking machinery that facilitates root hair developmental phase transitions and responses to changes in the root microenvironment.
Collapse
Affiliation(s)
- Cheol-Min Yoo
- Noble Research Institute, LLC, 2510 Sam Noble Parkway, Ardmore, OK, 73401, USA
- Present address: Gulf Coast Research and Education Center, University of Florida, 14625 CR 672, Wimauma, FL 33598, USA
| | - Satoshi Naramoto
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, 2-1-1, Katahira, Aobaku, Japan
| | - J. Alan Sparks
- Noble Research Institute, LLC, 2510 Sam Noble Parkway, Ardmore, OK, 73401, USA
| | - Bibi Rafeiza Khan
- Noble Research Institute, LLC, 2510 Sam Noble Parkway, Ardmore, OK, 73401, USA
| | - Jin Nakashima
- Noble Research Institute, LLC, 2510 Sam Noble Parkway, Ardmore, OK, 73401, USA
| | - Hiroo Fukuda
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | |
Collapse
|
35
|
Abstract
Members of the ADP-ribosylation factor (Arf) family of small GTP-binding (G) proteins regulate several aspects of membrane trafficking, such as vesicle budding, tethering and cytoskeleton organization. Arf family members, including Arf-like (Arl) proteins have been implicated in several essential cellular functions, like cell spreading and migration. These functions are used by cancer cells to disseminate and invade the tissues surrounding the primary tumor, leading to the formation of metastases. Indeed, Arf and Arl proteins, as well as their guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs) have been found to be abnormally expressed in different cancer cell types and human cancers. Here, we review the current evidence supporting the involvement of Arf family proteins and their GEFs and GAPs in cancer progression, focusing on 3 different mechanisms: cell-cell adhesion, integrin internalization and recycling, and actin cytoskeleton remodeling.
Collapse
Affiliation(s)
- Cristina Casalou
- a CEDOC, NOVA Medical School - Faculdade de Ciências Médicas, Universidade NOVA de Lisboa , Lisbon , Portugal
| | - Alexandra Faustino
- a CEDOC, NOVA Medical School - Faculdade de Ciências Médicas, Universidade NOVA de Lisboa , Lisbon , Portugal.,b ProRegeM PhD Program, NOVA Medical School - Faculdade de Ciências Médicas, Universidade NOVA de Lisboa , Lisbon , Portugal
| | - Duarte C Barral
- a CEDOC, NOVA Medical School - Faculdade de Ciências Médicas, Universidade NOVA de Lisboa , Lisbon , Portugal
| |
Collapse
|
36
|
Luo R, Chen PW, Wagenbach M, Jian X, Jenkins L, Wordeman L, Randazzo PA. Direct Functional Interaction of the Kinesin-13 Family Member Kinesin-like Protein 2A (Kif2A) and Arf GAP with GTP-binding Protein-like, Ankyrin Repeats and PH Domains1 (AGAP1). J Biol Chem 2016; 291:21350-21362. [PMID: 27531749 DOI: 10.1074/jbc.m116.732479] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 08/09/2016] [Indexed: 11/06/2022] Open
Abstract
The molecular basis for control of the cytoskeleton by the Arf GTPase-activating protein AGAP1 has not been characterized. AGAP1 is composed of G-protein-like (GLD), pleckstrin homology (PH), Arf GAP, and ankyrin repeat domains. Kif2A was identified in screens for proteins that bind to AGAP1. The GLD and PH domains of AGAP1 bound the motor domain of Kif2A. Kif2A increased GAP activity of AGAP1, and a protein composed of the GLD and PH domains of AGAP1 increased ATPase activity of Kif2A. Knockdown (KD) of Kif2A or AGAP1 slowed cell migration and accelerated cell spreading. The effect of Kif2A KD on spreading could be rescued by expression of Kif2A-GFP or FLAG-AGAP1, but not by Kif2C-GFP. The effect of AGAP1 KD could be rescued by FLAG-AGAP1, but not by an AGAP1 mutant that did not bind Kif2A efficiently, ArfGAP1-HA or Kif2A-GFP. Taken together, the results support the hypothesis that the Kif2A·AGAP1 complex contributes to control of cytoskeleton remodeling involved in cell movement.
Collapse
Affiliation(s)
- Ruibai Luo
- From the Laboratory of Cellular and Molecular Biology and
| | - Pei-Wen Chen
- From the Laboratory of Cellular and Molecular Biology and.,the Department of Biology, Grinnell College, Grinnell, Iowa 50112, and
| | - Michael Wagenbach
- the Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, Washington 98195
| | - Xiaoying Jian
- From the Laboratory of Cellular and Molecular Biology and
| | - Lisa Jenkins
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Linda Wordeman
- the Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, Washington 98195
| | | |
Collapse
|
37
|
Timbers TA, Garland SJ, Mohan S, Flibotte S, Edgley M, Muncaster Q, Au V, Li-Leger E, Rosell FI, Cai J, Rademakers S, Jansen G, Moerman DG, Leroux MR. Accelerating Gene Discovery by Phenotyping Whole-Genome Sequenced Multi-mutation Strains and Using the Sequence Kernel Association Test (SKAT). PLoS Genet 2016; 12:e1006235. [PMID: 27508411 PMCID: PMC4980031 DOI: 10.1371/journal.pgen.1006235] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 07/11/2016] [Indexed: 11/25/2022] Open
Abstract
Forward genetic screens represent powerful, unbiased approaches to uncover novel components in any biological process. Such screens suffer from a major bottleneck, however, namely the cloning of corresponding genes causing the phenotypic variation. Reverse genetic screens have been employed as a way to circumvent this issue, but can often be limited in scope. Here we demonstrate an innovative approach to gene discovery. Using C. elegans as a model system, we used a whole-genome sequenced multi-mutation library, from the Million Mutation Project, together with the Sequence Kernel Association Test (SKAT), to rapidly screen for and identify genes associated with a phenotype of interest, namely defects in dye-filling of ciliated sensory neurons. Such anomalies in dye-filling are often associated with the disruption of cilia, organelles which in humans are implicated in sensory physiology (including vision, smell and hearing), development and disease. Beyond identifying several well characterised dye-filling genes, our approach uncovered three genes not previously linked to ciliated sensory neuron development or function. From these putative novel dye-filling genes, we confirmed the involvement of BGNT-1.1 in ciliated sensory neuron function and morphogenesis. BGNT-1.1 functions at the trans-Golgi network of sheath cells (glia) to influence dye-filling and cilium length, in a cell non-autonomous manner. Notably, BGNT-1.1 is the orthologue of human B3GNT1/B4GAT1, a glycosyltransferase associated with Walker-Warburg syndrome (WWS). WWS is a multigenic disorder characterised by muscular dystrophy as well as brain and eye anomalies. Together, our work unveils an effective and innovative approach to gene discovery, and provides the first evidence that B3GNT1-associated Walker-Warburg syndrome may be considered a ciliopathy. Model organisms are useful tools for uncovering new genes involved in a biological process via genetic screens. Such an approach is powerful, but suffers from drawbacks that can slow down gene discovery. In forward genetics screens, difficult-to-map phenotypes present daunting challenges, and whole-genome coverage can be equally challenging for reverse genetic screens where typically only a single gene’s function is assayed per strain. Here, we show a different approach which includes positive aspects of forward (high-coverage, randomly-induced mutations) and reverse genetics (prior knowledge of gene disruption) to accelerate gene discovery. We paired a whole-genome sequenced multi-mutation C. elegans library with a rare-variant associated test to rapidly identify genes associated with a phenotype of interest: defects in sensory neurons bearing sensory organelles called cilia, via a simple dye-filling assay to probe the form and function of these cells. We found two well characterised dye-filling genes and three genes, not previously linked to ciliated sensory neuron development or function, that were associated with dye-filling defects. We reveal that disruption of one of these (BGNT-1.1), whose human orthologue is associated with Walker-Warburg syndrome, results in abrogated uptake of dye and cilia length defects. We believe that our novel approach is useful for any organism with a small genome that can be quickly sequenced and where many mutant strains can be easily isolated and phenotyped, such as Drosophila and Arabidopsis.
Collapse
Affiliation(s)
- Tiffany A. Timbers
- Department of Molecular Biology and Biochemistry and Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Stephanie J. Garland
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Swetha Mohan
- Department of Molecular Biology and Biochemistry and Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Stephane Flibotte
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mark Edgley
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Quintin Muncaster
- Department of Molecular Biology and Biochemistry and Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Vinci Au
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Erica Li-Leger
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Federico I. Rosell
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jerry Cai
- Department of Molecular Biology and Biochemistry and Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | - Gert Jansen
- Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | - Donald G. Moerman
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michel R. Leroux
- Department of Molecular Biology and Biochemistry and Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
- * E-mail:
| |
Collapse
|
38
|
ACAP3 regulates neurite outgrowth through its GAP activity specific to Arf6 in mouse hippocampal neurons. Biochem J 2016; 473:2591-602. [PMID: 27330119 DOI: 10.1042/bcj20160183] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/21/2016] [Indexed: 01/30/2023]
Abstract
ACAP3 (ArfGAP with coiled-coil, ankyrin repeat and pleckstrin homology domains 3) belongs to the ACAP family of GAPs (GTPase-activating proteins) for the small GTPase Arf (ADP-ribosylation factor). However, its specificity to Arf isoforms and physiological functions remain unclear. In the present study, we demonstrate that ACAP3 plays an important role in neurite outgrowth of mouse hippocampal neurons through its GAP activity specific to Arf6. In primary cultured mouse hippocampal neurons, knockdown of ACAP3 abrogated neurite outgrowth, which was rescued by ectopically expressed wild-type ACAP3, but not by its GAP activity-deficient mutant. Ectopically expressed ACAP3 in HEK (human embryonic kidney)-293T cells showed the GAP activity specific to Arf6. In support of this observation, the level of GTP-bound Arf6 was significantly increased by knockdown of ACAP3 in hippocampal neurons. In addition, knockdown and knockout of Arf6 in mouse hippocampal neurons suppressed neurite outgrowth. These results demonstrate that ACAP3 positively regulates neurite outgrowth through its GAP activity specific to Arf6. Furthermore, neurite outgrowth suppressed by ACAP3 knockdown was rescued by expression of a fast cycle mutant of Arf6 that spontaneously exchanges guanine nucleotides on Arf6, but not by that of wild-type, GTP- or GDP-locked mutant Arf6. Thus cycling between active and inactive forms of Arf6, which is precisely regulated by ACAP3 in concert with a guanine-nucleotide-exchange factor(s), seems to be required for neurite outgrowth of hippocampal neurons.
Collapse
|
39
|
Fukaya M, Ohta S, Hara Y, Tamaki H, Sakagami H. Distinct subcellular localization of alternative splicing variants of EFA6D, a guanine nucleotide exchange factor for Arf6, in the mouse brain. J Comp Neurol 2016; 524:2531-52. [PMID: 27241101 DOI: 10.1002/cne.24048] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 03/31/2016] [Accepted: 05/24/2016] [Indexed: 11/07/2022]
Abstract
EFA6D (guanine nucleotide exchange factor for ADP-ribosylation factor 6 [Arf6]D) is also known as EFA6R, Psd3, and HCA67. It is the fourth member of the EFA6 family with guanine nucleotide exchange activity for Arf6, a small guanosine triphosphatase (GTPase) that regulates endosomal trafficking and actin cytoskeleton remodeling. We propose a classification and nomenclature of 10 EFA6D variants deposited in the GenBank database as EFA6D1a, 1b, 1c, 1d, 1s, 2a, 2b, 2c, 2d, and 2s based on the combination of N-terminal and C-terminal insertions. Polymerase chain reaction analysis showed the expression of all EFA6D variants except for variants a and d in the adult mouse brain. Immunoblotting analysis with novel variant-specific antibodies showed the endogenous expression of EFA6D1b, EFA6D1c, and EFA6D1s at the protein level, with the highest expression being EFA6D1s, in the brain. Immunoblotting analysis of forebrain subcellular fractions showed the distinct subcellular distribution of EFA6D1b/c and EFA6D1s. The immunohistochemical analysis revealed distinct but overlapping immunoreactive patterns between EFA6D1b/c and EFA6D1s in the mouse brain. In immunoelectron microscopic analyses of the hippocampal CA3 region, EFA6D1b/c was present predominantly in the mossy fiber axons of dentate granule cells, whereas EFA6D1s was present abundantly in the cell bodies, dendritic shafts, and spines of hippocampal pyramidal cells. These results provide the first anatomical evidence suggesting the functional diversity of EFA6D variants, particularly EFA6D1b/c and EFA6D1s, in neurons. J. Comp. Neurol. 524:2531-2552, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Masahiro Fukaya
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Shingo Ohta
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yoshinobu Hara
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Hideaki Tamaki
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Hiroyuki Sakagami
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| |
Collapse
|
40
|
Mrozowska PS, Fukuda M. Regulation of podocalyxin trafficking by Rab small GTPases in 2D and 3D epithelial cell cultures. J Cell Biol 2016; 213:355-69. [PMID: 27138252 PMCID: PMC4862332 DOI: 10.1083/jcb.201512024] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 03/18/2016] [Indexed: 11/22/2022] Open
Abstract
MDCK II cells, a widely used model of polarized epithelia, develop into different structures depending on culture conditions: two-dimensional (2D) monolayers when grown on synthetic supports or three-dimensional (3D) cysts when surrounded by an extracellular matrix. The establishment of epithelial polarity is accompanied by transcytosis of the apical marker podocalyxin from the outer plasma membrane to the newly formed apical domain, but its exact route and regulation remain poorly understood. Here, through comprehensive colocalization and knockdown screenings, we identified the Rab GTPases mediating podocalyxin transcytosis and showed that different sets of Rabs coordinate its transport during cell polarization in 2D and 3D structures. Moreover, we demonstrated that different Rab35 effectors regulate podocalyxin trafficking in 2D and 3D environments; trafficking is mediated by OCRL in 2D monolayers and ACAP2 in 3D cysts. Our results give substantial insight into regulation of the transcytosis of this apical marker and highlight differences between trafficking mechanisms in 2D and 3D cell cultures.
Collapse
Affiliation(s)
- Paulina S Mrozowska
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
41
|
Roy NS, Yohe ME, Randazzo PA, Gruschus JM. Allosteric properties of PH domains in Arf regulatory proteins. CELLULAR LOGISTICS 2016; 6:e1181700. [PMID: 27294009 DOI: 10.1080/21592799.2016.1181700] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 10/21/2022]
Abstract
Pleckstrin Homology (PH) domains bind phospholipids and proteins. They are critical regulatory elements of a number enzymes including guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs) for Ras-superfamily guanine nucleotide binding proteins such as ADP-ribosylation factors (Arfs). Recent studies have indicated that many PH domains may bind more than one ligand cooperatively. Here we discuss the molecular basis of PH domain-dependent allosteric behavior of 2 ADP-ribosylation factor exchange factors, Grp1 and Brag2, cooperative binding of ligands to the PH domains of Grp1 and the Arf GTPase-activating protein, ASAP1, and the consequences for activity of the associated catalytic domains.
Collapse
Affiliation(s)
- Neeladri Sekhar Roy
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health , Bethesda, MD, USA
| | - Marielle E Yohe
- Genetics Branch, National Cancer Institute, National Institutes of Health , Bethesda, MD, USA
| | - Paul A Randazzo
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health , Bethesda, MD, USA
| | - James M Gruschus
- Laboratory of Structural Biophysics, National Heart, Lung and Blood Institute, National Institutes of Health , Bethesda, MD, USA
| |
Collapse
|
42
|
Naramoto S, Dainobu T, Tokunaga H, Kyozuka J, Fukuda H. Cellular and developmental function of ACAP type ARF-GAP proteins are diverged in plant cells. PLANT BIOTECHNOLOGY (TOKYO, JAPAN) 2016; 33:309-314. [PMID: 31274992 PMCID: PMC6565945 DOI: 10.5511/plantbiotechnology.16.0309a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 03/09/2016] [Indexed: 05/29/2023]
Abstract
Vesicle transport is crucial for various cellular functions and development of multicellular organisms. ARF-GAP is one of the key regulators of vesicle transport and is diverse family of proteins. Arabidopsis has 15 ARF-GAP proteins and four members are classified as ACAP type ARF-GAP proteins. Our previous study identified that VASCULAR NETWORK DEFECTIVE3 (VAN3), an ACAP ARF-GAP, played crucial roles in leaf vascular formation. However, it remains question how other members of plant ACAP ARF-GAPs function in cellular and developmental processes. To characterize these, we analyzed spatial expression pattern and subcellular localization of VAN3 and three other ACAPs, so called VAN3-like proteins (VALs). Expression pattern analysis revealed that they were expressed in distinctive developmental processes. Subcellular localization analysis in protoplast cells indicated that in contrast to VAN3, which localizes on trans-Golgi networks/early endosomes (TGNs/EEs), VAL1 and VAL2 were localized on ARA6-labelled endosomes, and VAL3 resided mainly in the cytoplasm. These results indicated that VAN3 and VALs are differently expressed in a tissue level and function in different intracellular compartments, in spite of their significant sequence similarities. These findings suggested functional divergence among plant ACAPs. Cellular localizations of all members of animal ACAP proteins are identical. Therefore our findings also suggested that plant evolved ACAP proteins in plant specific manner.
Collapse
Affiliation(s)
- Satoshi Naramoto
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8577, Japan
| | - Tomoko Dainobu
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8577, Japan
| | - Hiroki Tokunaga
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8577, Japan
| | - Junko Kyozuka
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8577, Japan
| | - Hiroo Fukuda
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
43
|
Sullivan KD, Nakagawa A, Xue D, Espinosa JM. Human ACAP2 is a homolog of C. elegans CNT-1 that promotes apoptosis in cancer cells. Cell Cycle 2016; 14:1771-8. [PMID: 25853217 DOI: 10.1080/15384101.2015.1026518] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Activation of caspases is an integral part of the apoptotic cell death program. Collectively, these proteases target hundreds of substrates, leading to the hypothesis that apoptosis is "death by a thousand cuts". Recent work, however, has demonstrated that caspase cleavage of only a subset of these substrates directs apoptosis in the cell. One such example is C. elegans CNT-1, which is cleaved by CED-3 to generate a truncated form, tCNT-1, that acquires a potent phosphoinositide-binding activity and translocates to the plasma membrane where it inactivates AKT survival signaling. We report here that ACAP2, a homolog of C. elegans CNT-1, has a pro-apoptotic function and an identical phosphoinositide-binding pattern to that of tCNT-1, despite not being an apparent target of caspase cleavage. We show that knockdown of ACAP2 blocks apoptosis in cancer cells in response to the chemotherapeutic antimetabolite 5-fluorouracil and that ACAP2 expression is down-regulated in some esophageal cancers, leukemias and lymphomas. These results suggest that ACAP2 is a functional homolog of C. elegans CNT-1 and its inactivation or downregulation in human cells may contribute to cancer development.
Collapse
Affiliation(s)
- Kelly D Sullivan
- a Department of Molecular, Cellular, and Developmental Biology; University of Colorado ; Boulder , CO , USA
| | | | | | | |
Collapse
|
44
|
Molecular imaging analysis of Rab GTPases in the regulation of phagocytosis and macropinocytosis. Anat Sci Int 2015; 91:35-42. [DOI: 10.1007/s12565-015-0313-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/26/2015] [Indexed: 12/22/2022]
|
45
|
Regulators and Effectors of Arf GTPases in Neutrophils. J Immunol Res 2015; 2015:235170. [PMID: 26609537 PMCID: PMC4644846 DOI: 10.1155/2015/235170] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 09/30/2015] [Indexed: 12/22/2022] Open
Abstract
Polymorphonuclear neutrophils (PMNs) are key innate immune cells that represent the first line of defence against infection. They are the first leukocytes to migrate from the blood to injured or infected sites. This process involves molecular mechanisms that coordinate cell polarization, delivery of receptors, and activation of integrins at the leading edge of migrating PMNs. These phagocytes actively engulf microorganisms or form neutrophil extracellular traps (NETs) to trap and kill pathogens with bactericidal compounds. Association of the NADPH oxidase complex at the phagosomal membrane for production of reactive oxygen species (ROS) and delivery of proteolytic enzymes into the phagosome initiate pathogen killing and removal. G protein-dependent signalling pathways tightly control PMN functions. In this review, we will focus on the small monomeric GTPases of the Arf family and their guanine exchange factors (GEFs) and GTPase activating proteins (GAPs) as components of signalling cascades regulating PMN responses. GEFs and GAPs are multidomain proteins that control cellular events in time and space through interaction with other proteins and lipids inside the cells. The number of Arf GAPs identified in PMNs is expanding, and dissecting their functions will provide important insights into the role of these proteins in PMN physiology.
Collapse
|
46
|
Activation-Inactivation Cycling of Rab35 and ARF6 Is Required for Phagocytosis of Zymosan in RAW264 Macrophages. J Immunol Res 2015; 2015:429439. [PMID: 26229970 PMCID: PMC4502309 DOI: 10.1155/2015/429439] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 05/25/2015] [Indexed: 12/26/2022] Open
Abstract
Phagocytosis of zymosan by phagocytes is a widely used model of microbial recognition by the innate immune system. Live-cell imaging showed that fluorescent protein-fused Rab35 accumulated in the membranes of phagocytic cups and then dissociated from the membranes of newly formed phagosomes. By our novel pull-down assay for Rab35 activity, we found that Rab35 is deactivated immediately after zymosan internalization into the cells. Phagosome formation was inhibited in cells expressing the GDP- or GTP-locked Rab35 mutant. Moreover, the simultaneous expression of ACAP2-a Rab35 effector protein-with GTP-locked Rab35 or the expression of plasma membrane-targeted ACAP2 showed a marked inhibitory effect on phagocytosis through ARF6 inactivation by the GAP activity of ACAP2. ARF6, a substrate for ACAP2, was also localized on the phagocytic cups and dissociated from the membranes of internalized phagosomes. In support of the microscopic observations, ARF6-GTP pull-down experiments showed that ARF6 is transiently activated during phagosome formation. Furthermore, the expression of GDP- or GTP-locked ARF6 mutants also suppresses the uptake of zymosan. These data suggest that the activation-inactivation cycles of Rab35 and ARF6 are required for the uptake of zymosan and that ACAP2 is an important component that links Rab35/ARF6 signaling during phagocytosis of zymosan.
Collapse
|
47
|
Dutta D, Donaldson JG. Sorting of Clathrin-Independent Cargo Proteins Depends on Rab35 Delivered by Clathrin-Mediated Endocytosis. Traffic 2015; 16:994-1009. [PMID: 25988331 DOI: 10.1111/tra.12302] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 05/14/2015] [Accepted: 05/14/2015] [Indexed: 01/13/2023]
Abstract
Clathrin-mediated endocytosis (CME) and clathrin-independent endocytosis (CIE) co-exist in most cells but little is known about their communication and coordination. Here we show that when CME was inhibited, endocytosis by CIE continued but endosomal trafficking of CIE cargo proteins was altered. CIE cargo proteins that normally traffic directly into Arf6-associated tubules after internalization and avoid degradation (CD44, CD98 and CD147) now trafficked to lysosomes and were degraded. The endosomal tubules were also absent and Arf6-GTP levels were elevated. The altered trafficking, loss of the tubular endosomal network and elevated Arf6-GTP levels caused by inhibition of CME were rescued by expression of Rab35, a Rab associated with clathrin-coated vesicles, or its effector ACAPs, Arf6 GTPase activating proteins (GAP) that inactivate Arf6. Furthermore, siRNA knockdown of Rab35 recreated the phenotype of CME ablation on CIE cargo trafficking without altering endocytosis of transferrin. These observations suggest that Rab35 serves as a CME detector and that loss of CME, or Rab35 input, leads to elevated Arf6-GTP and shifts the sorting of CIE cargo proteins to lysosomes and degradation.
Collapse
Affiliation(s)
- Dipannita Dutta
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Julie G Donaldson
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
48
|
Etoh K, Fukuda M. Structure-function analyses of the small GTPase Rab35 and its effector protein centaurin-β2/ACAP2 during neurite outgrowth of PC12 cells. J Biol Chem 2015; 290:9064-74. [PMID: 25694427 DOI: 10.1074/jbc.m114.611301] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Indexed: 11/06/2022] Open
Abstract
The small GTPase Rab35 is a molecular switch for membrane trafficking that regulates a variety of cellular events. We previously showed that Rab35 promotes neurite outgrowth of nerve growth factor-stimulated PC12 cells through interaction with centaurin-β2 (also called ACAP2). Centaurin-β2 is the only Rab35-binding protein reported thus far that exclusively recognizes Rab35 and does not recognize any of the other 59 Rabs identified in mammals, but the molecular basis for the exclusive specificity of centaurin-β2 for Rab35 has remained completely unknown. In this study, we performed deletion and mutation analyses and succeeded in identifying the residues of Rab35 and centaurin-β2 that are crucial for formation of a Rab35·centaurin-β2 complex. We found that two threonine residues (Thr-76 and Thr-81) in the switch II region of Rab35 are responsible for binding centaurin-β2 and that the same residues are dispensable for Rab35 recognition by other Rab35-binding proteins. We also determined the minimal Rab35-binding site of centaurin-β2 and identified two asparagine residues (Asn-610 and Asn-691) in the Rab35-binding site as key residues for its specific Rab35 recognition. We further showed by knockdown-rescue approaches that neither a centaurin-β2 binding-deficient Rab35(T76S/T81A) mutant nor a Rab35 binding-deficient centaurin-β2(N610A/N691A) mutant supported neurite outgrowth of PC12 cells, thereby demonstrating the functional significance of the Rab35/centaurin-β2 interaction during neurite outgrowth of PC12 cells.
Collapse
Affiliation(s)
- Kan Etoh
- From the Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Mitsunori Fukuda
- From the Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
49
|
Davidson AC, Humphreys D, Brooks ABE, Hume PJ, Koronakis V. The Arf GTPase-activating protein family is exploited by Salmonella enterica serovar Typhimurium to invade nonphagocytic host cells. mBio 2015; 6:e02253-14. [PMID: 25670778 PMCID: PMC4337568 DOI: 10.1128/mbio.02253-14] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 12/05/2014] [Indexed: 01/06/2023] Open
Abstract
UNLABELLED To establish intracellular infections, Salmonella bacteria trigger host cell membrane ruffling and invasion by subverting cellular Arf guanine nucleotide exchange factors (GEFs) that activate Arf1 and Arf6 GTPases by promoting GTP binding. A family of cellular Arf GTPase-activating proteins (GAPs) can downregulate Arf signaling by stimulating GTP hydrolysis, but whether they do this during infection is unknown. Here, we uncovered a remarkable role for distinct Arf GAP family members in Salmonella invasion. The Arf6 GAPs ACAP1 and ADAP1 and the Arf1 GAP ASAP1 localized at Salmonella-induced ruffles, which was not the case for the plasma membrane-localized Arf6 GAPs ARAP3 and GIT1 or the Golgi-associated Arf1 GAP1. Surprisingly, we found that loss of ACAP1, ADAP1, or ASAP1 impaired Salmonella invasion, revealing that GAPs cannot be considered mere terminators of cytoskeleton remodeling. Salmonella invasion was restored in Arf GAP-depleted cells by expressing fast-cycling Arf derivatives, demonstrating that Arf GTP/GDP cycles facilitate Salmonella invasion. Consistent with this view, both constitutively active and dominant-negative Arf derivatives that cannot undergo GTP/GDP cycles inhibited invasion. Furthermore, we demonstrated that Arf GEFs and GAPs colocalize at invading Salmonella and collaborate to drive Arf1-dependent pathogen invasion. This study revealed that Salmonella bacteria exploit a remarkable interplay between Arf GEFs and GAPs to direct cycles of Arf GTPase activation and inactivation. These cycles drive Salmonella cytoskeleton remodeling and enable intracellular infections. IMPORTANCE To initiate infections, the Salmonella bacterial pathogen remodels the mammalian actin cytoskeleton and invades host cells by subverting host Arf GEFs that activate Arf1 and Arf6 GTPases. Cellular Arf GAPs deactivate Arf GTPases and negatively regulate cell processes, but whether they target Arfs during infection is unknown. Here, we uncovered an important role for the Arf GAP family in Salmonella invasion. Surprisingly, we found that Arf1 and Arf6 GAPs cooperate with their Arf GEF counterparts to facilitate cycles of Arf GTPase activation and inactivation, which direct pathogen invasion. This report illustrates that GAP proteins promote actin-dependent processes and are not necessarily restricted to negatively regulating cellular signaling. It uncovers a remarkable interplay between Arf GEFs and GAPs that is exploited by Salmonella to establish infection and expands our understanding of Arf GTPase-regulated cytoskeleton remodeling.
Collapse
Affiliation(s)
- Anthony C Davidson
- Department of Pathology, Cambridge University, Cambridge, United Kingdom
| | - Daniel Humphreys
- Department of Pathology, Cambridge University, Cambridge, United Kingdom
| | - Andrew B E Brooks
- Department of Pathology, Cambridge University, Cambridge, United Kingdom
| | - Peter J Hume
- Department of Pathology, Cambridge University, Cambridge, United Kingdom
| | - Vassilis Koronakis
- Department of Pathology, Cambridge University, Cambridge, United Kingdom
| |
Collapse
|
50
|
Pang X, Fan J, Zhang Y, Zhang K, Gao B, Ma J, Li J, Deng Y, Zhou Q, Egelman EH, Hsu VW, Sun F. A PH domain in ACAP1 possesses key features of the BAR domain in promoting membrane curvature. Dev Cell 2014; 31:73-86. [PMID: 25284369 PMCID: PMC4198613 DOI: 10.1016/j.devcel.2014.08.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 08/12/2014] [Accepted: 08/21/2014] [Indexed: 11/29/2022]
Abstract
The BAR (Bin-Amphiphysin-Rvs) domain undergoes dimerization to produce a curved protein structure, which superimposes onto membrane through electrostatic interactions to sense and impart membrane curvature. In some cases, a BAR domain also possesses an amphipathic helix that inserts into the membrane to induce curvature. ACAP1 (Arfgap with Coil coil, Ankyrin repeat, and PH domain protein 1) contains a BAR domain. Here, we show that this BAR domain can neither bind membrane nor impart curvature, but instead requires a neighboring PH (Pleckstrin Homology) domain to achieve these functions. Specific residues within the PH domain are responsible for both membrane binding and curvature generation. The BAR domain adjacent to the PH domain instead interacts with the BAR domains of neighboring ACAP1 proteins to enable clustering at the membrane. Thus, we have uncovered the molecular basis for an unexpected and unconventional collaboration between PH and BAR domains in membrane bending.
Collapse
Affiliation(s)
- Xiaoyun Pang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Fan
- Department of Physics and Materials Science, City University of Hong Kong, Hong Kong 999077, China
| | - Yan Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Kai Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Bingquan Gao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Ma
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jian Li
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Yuchen Deng
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiangjun Zhou
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Edward H Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - Victor W Hsu
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| | - Fei Sun
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Center for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|