1
|
Nikhil K, Shah K. The significant others of aurora kinase a in cancer: combination is the key. Biomark Res 2024; 12:109. [PMID: 39334449 PMCID: PMC11438406 DOI: 10.1186/s40364-024-00651-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
AURKA is predominantly famous as an essential mitotic kinase. Recent findings have also established its critical role in a plethora of other biological processes including ciliogenesis, mitochondrial dynamics, neuronal outgrowth, DNA replication and cell cycle progression. AURKA overexpression in numerous cancers is strongly associated with poor prognosis and survival. Still no AURKA-targeted drug has been approved yet, partially because of the associated collateral toxicity and partly due to its limited efficacy as a single agent in a wide range of tumors. Mechanistically, AURKA overexpression allows it to phosphorylate numerous pathological substrates promoting highly aggressive oncogenic phenotypes. Our review examines the most recent advances in AURKA regulation and focuses on 33 such direct cancer-specific targets of AURKA and their associated oncogenic signaling cascades. One of the common themes that emerge is that AURKA is often involved in a feedback loop with its substrates, which could be the decisive factor causing its sustained upregulation and hyperactivation in cancer cells, an Achilles heel not exploited before. This dynamic interplay between AURKA and its substrates offers potential opportunities for targeted therapeutic interventions. By targeting these substrates, it may be possible to disrupt this feedback loop to effectively reverse AURKA levels, thereby providing a promising avenue for developing safer AURKA-targeted therapeutics. Additionally, exploring the synergistic effects of AURKA inhibition with its other oncogenic and/or tumor-suppressor targets could provide further opportunities for developing effective combination therapies against AURKA-driven cancers, thereby maximizing its potential as a critical drug target.
Collapse
Affiliation(s)
- Kumar Nikhil
- Department of Chemistry, Purdue University Institute for Cancer Research, 560 Oval Drive, West Lafayette, IN, 47907, USA.
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, 751024, India.
| | - Kavita Shah
- Department of Chemistry, Purdue University Institute for Cancer Research, 560 Oval Drive, West Lafayette, IN, 47907, USA.
| |
Collapse
|
2
|
Thulasidharan A, Garg L, Tendulkar S, Ratnaparkhi GS. Age-dependent dynamics of neuronal VAPB ALS inclusions in the adult brain. Neurobiol Dis 2024; 196:106517. [PMID: 38679111 DOI: 10.1016/j.nbd.2024.106517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/01/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a relentlessly progressive and fatal disease, caused by the degeneration of upper and lower motor neurons within the brain and spinal cord in the ageing human. The dying neurons contain cytoplasmic inclusions linked to the onset and progression of the disease. Here, we use a Drosophila model of ALS8 (VAPP58S) to understand the modulation of these inclusions in the ageing adult brain. The adult VAPP58S fly shows progressive deterioration in motor function till its demise 25 days post-eclosion. The density of VAPP58S-positive brain inclusions is stable for 5-15 days of age. In contrast, adding a single copy of VAPWT to the VAPP58S animal leads to a large decrease in inclusion density with concomitant rescue of motor function and lifespan. ER stress, a contributing factor in disease, shows reduction with ageing for the disease model. Autophagy, rather than the Ubiquitin Proteasome system, is the dominant mechanism for aggregate clearance. We explored the ability of Drosophila Valosin-containing protein (VCP/TER94), the ALS14 locus, which is involved in cellular protein clearance, to regulate age-dependent aggregation. Contrary to expectation, TER94 overexpression increased VAPP58S punctae density, while its knockdown led to enhanced clearance. Expression of a dominant positive allele, TER94R152H, further stabilised VAPP58S puncta, cementing roles for an ALS8-ALS14 axis. Our results are explained by a mechanism where autophagy is modulated by TER94 knockdown. Our study sheds light on the complex regulatory events involved in the neuronal maintenance of ALS8 aggregates, suggesting a context-dependent switch between proteasomal and autophagy-based mechanisms as the larvae develop into an adult. A deeper understanding of the nucleation and clearance of the inclusions, which affect cellular stress and function, is essential for understanding the initiation and progression of ALS.
Collapse
Affiliation(s)
- Aparna Thulasidharan
- Department of Biology, Indian Institute of Science Education & Research (IISER), Pune 411008, India
| | - Lovleen Garg
- Department of Biology, Indian Institute of Science Education & Research (IISER), Pune 411008, India
| | - Shweta Tendulkar
- Department of Biology, Indian Institute of Science Education & Research (IISER), Pune 411008, India
| | - Girish S Ratnaparkhi
- Department of Biology, Indian Institute of Science Education & Research (IISER), Pune 411008, India.
| |
Collapse
|
3
|
Su Y, Zhao L, Lei D, Yang X. Inhibition of circ_0073932 attenuates myocardial ischemia‒reperfusion injury via miR-493-3p/FAF1/JNK. In Vitro Cell Dev Biol Anim 2024; 60:628-643. [PMID: 38578382 DOI: 10.1007/s11626-024-00900-8] [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: 09/22/2023] [Accepted: 02/29/2024] [Indexed: 04/06/2024]
Abstract
Oxidative stress and apoptosis play crucial roles in myocardial ischemia‒reperfusion injury (MIRI). In this study, we investigated the role of circ_0073932 in MIRI as well as its molecular mechanism. A hypoxia/reoxygenation (H/R) cardiomyocyte model was established with H9C2 cardiomyocytes, and RT-qPCR was used to measure gene expression. We observed that circ_0073932 expression was abnormally increased in the H/R cardiomyocyte model and in blood samples from MIRI patients. Inhibition of circ_0073932 suppressed H/R-induced cell apoptosis, oxidative stress (ROS, LDH and MDA), and p-JNK expression. Dual luciferase reporter assays showed that circ_0073932 targeted the downregulation of miR-493-3p, and miR-493-3p targeted the downregulation of FAF1. Furthermore, si-circ_0073932, an miR-493-3p inhibitor, oe-FAF1, or si-FAF1 were transfected into H9C2 cardiomyocytes to investigate the roles of these factors in MIRI. Our results showed that compared with the H/R group, si-circ_0073932 inhibited H/R-induced cell apoptosis, oxidative stress (ROS, LDH and MDA), and p-JNK expression. These results were reversed by the miR-493-3p inhibitor or oe-FAF1. Finally, a rat model of MIRI was established, and si-circ_0073932 was administered. Inhibition of circ_0073932 reduced the area of myocardial infarction and decreased the levels of apoptosis and oxidative stress by inhibiting the JNK signaling pathway. Our study indicated that circ_0073932 mediates MIRI via miR-493-3p/FAF1/JNK in vivo and in vitro, revealing novel insights into the pathogenesis of MIRI and providing a new target for the clinical treatment of MIRI.
Collapse
Affiliation(s)
- Yang Su
- The Outpatient Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Lili Zhao
- Radiology Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Dongli Lei
- Intensive Care Unit, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China
| | - Xiaoming Yang
- Information Statistics Centre, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, Yunnan, China.
| |
Collapse
|
4
|
Chen CH, Lin HW, Huang MF, Chiang CW, Lee KH, Phuong NT, Cai ZY, Chang WC, Lin DY. Sumoylation of SAP130 regulates its interaction with FAF1 as well as its protein stability and transcriptional repressor function. BMC Mol Cell Biol 2024; 25:2. [PMID: 38172660 PMCID: PMC10765799 DOI: 10.1186/s12860-023-00498-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Fas-associated factor 1 (FAF1) is a multidomain protein that interacts with diverse partners to affect numerous cellular processes. Previously, we discovered two Small Ubiquitin-like Modifier (SUMO)-interacting motifs (SIMs) within FAF1 that are crucial for transcriptional modulation of mineralocorticoid receptor. Recently, we identified Sin3A-associated protein 130 (SAP130), a putative sumoylated protein, as a candidate FAF1 interaction partner by yeast two-hybrid screening. However, it remained unclear whether SAP130 sumoylation might occur and functionally interact with FAF1. RESULTS In this study, we first show that SAP130 can be modified by SUMO1 at Lys residues 794, 878 and 932 both in vitro and in vivo. Mutation of these three SUMO-accepting Lys residues to Ala had no impact on SAP130 association with Sin3A or its nuclear localization, but the mutations abrogated the association of SAP130 with the FAF1. The mutations also potentiated SAP130 trans-repression activity and attenuated SAP130-mediated promotion of cell growth. Additionally, SUMO1-modified SAP130 was less stable than unmodified SAP130. Transient transfection experiments further revealed that FAF1 mitigated the trans-repression and cell proliferation-promoting functions of SAP130, and promoted SAP130 degradation by enhancing its polyubiquitination in a sumoylation-dependent manner. CONCLUSIONS Together, these results demonstrate that sumoylation of SAP130 regulates its biological functions and that FAF1 plays a crucial role in controlling the SUMO-dependent regulation of transcriptional activity and protein stability of SAP130.
Collapse
Affiliation(s)
- Chang-Han Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, 407219, Taiwan, ROC
- Department of Applied Chemistry, and Graduate Institute of Biomedicine and Biomedical Technology, National Chi Nan University, Nantou, 545301, Taiwan, ROC
| | - Hung-Wei Lin
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan, ROC
| | - Meng-Fang Huang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan, ROC
| | - Chi-Wu Chiang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC
| | - Kuen-Haur Lee
- Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan, ROC
| | - Nguyen Thanh Phuong
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan, ROC
| | - Zong-Yan Cai
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan, ROC
| | - Wen-Chang Chang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei Medical University, Taipei, 11031, Taiwan, ROC
| | - Ding-Yen Lin
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan, ROC.
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC.
| |
Collapse
|
5
|
Luo Z, Qin YK, Zhao K, Nan XY, Li WW, Li EC, Wang Q. Caspar negatively regulates anti-bacterial immunity by controlling the nuclear translocation of Relish in Chinese mitten crab (Eriocheir sinensis). FISH & SHELLFISH IMMUNOLOGY 2023; 136:108714. [PMID: 36990260 DOI: 10.1016/j.fsi.2023.108714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/23/2023] [Accepted: 03/26/2023] [Indexed: 06/19/2023]
Abstract
Caspar, a homolog of the Fas-associated factor 1 (FAF1) family, contains an N-terminal ubiquitin interaction domain, a ubiquitin-like self-association domain, and a C-terminal ubiquitin regulatory domain. Caspar has been reported to be involved in the antibacterial immunity of Drosophila, which is unclear whether it is involved in the antibacterial immune process of crustaceans. In this article, we identified a Caspar gene in Eriocheir sinensis and named it EsCaspar. EsCaspar positively respond to bacterial stimulation and downregulate the expression of certain associated antimicrobial peptides by inhibiting the nuclear translocation of EsRelish. Thus, EsCaspar might be a suppressor of the immune deficiency (IMD) pathway that prevents over-activation of the immune system. Indeed, excess EsCaspar protein in crabs reduced resistance to bacterial infection. In conclusion, EsCaspar is a suppressor of the IMD pathway in crabs that plays a negative regulatory role in antimicrobial immunity.
Collapse
Affiliation(s)
- Zhi Luo
- Laboratory of Immunological Defense, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Yu-Kai Qin
- Laboratory of Immunological Defense, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Ke Zhao
- Laboratory of Immunological Defense, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Xing-Yu Nan
- Laboratory of Immunological Defense, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Wei-Wei Li
- Laboratory of Immunological Defense, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Er-Chao Li
- Laboratory of Immunological Defense, School of Life Sciences, East China Normal University, 200241, Shanghai, China.
| | - Qun Wang
- Laboratory of Immunological Defense, School of Life Sciences, East China Normal University, 200241, Shanghai, China.
| |
Collapse
|
6
|
Cui S, Ye J. A protein-lipid complex that detoxifies free fatty acids. Bioessays 2023; 45:e2200210. [PMID: 36585363 PMCID: PMC9974861 DOI: 10.1002/bies.202200210] [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: 10/28/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 01/01/2023]
Abstract
Fatty acids (FAs) are well known to serve as substrates for reactions that provide cells with membranes and energy. In contrast to these metabolic reactions, the physiological importance of FAs themselves known as free FAs (FFAs) in cells remains obscure. Since accumulation of FFAs in cells is toxic, cells must develop mechanisms to detoxify FFAs. One such mechanism is to sequester free polyunsaturated FAs (PUFAs) into a droplet-like structure assembled by Fas-Associated Factor 1 (FAF1), a cytosolic protein. This sequestration limits access of PUFAs to Fe2+ , thereby preventing Fe2+ -catalyzed PUFA peroxidation. Consequently, assembly of the FAF1-FFA complex is critical to protect cells from ferroptosis, a cell death pathway triggered by PUFA peroxidation. The observations that free PUFAs in cytosol are not randomly diffused but rather sequestered into a membraneless complex should open new directions to explore signaling pathways by which FFAs regulate cellular physiology.
Collapse
Affiliation(s)
- Shaojie Cui
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jin Ye
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
7
|
Tendulkar S, Hegde S, Garg L, Thulasidharan A, Kaduskar B, Ratnaparkhi A, Ratnaparkhi GS. Caspar, an adapter for VAPB and TER94, modulates the progression of ALS8 by regulating IMD/NFκB mediated glial inflammation in a drosophila model of human disease. Hum Mol Genet 2022; 31:2857-2875. [PMID: 35377453 PMCID: PMC9433731 DOI: 10.1093/hmg/ddac076] [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: 01/10/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal, late-onset, progressive motor neurodegenerative disorder. A key pathological feature of the disease is the presence of heavily ubiquitinated protein inclusions. Both the unfolded protein response and the ubiquitin–proteasome system appear significantly impaired in patients and animal models of ALS. We have studied cellular and molecular mechanisms involved in ALS using a vesicle-associated membrane protein-associated protein B (VAPB/ALS8) Drosophila model [Moustaqim-Barrette, A., Lin, Y.Q., Pradhan, S., Neely, G.G., Bellen, H.J. and Tsuda, H. (2014) The ALS 8 protein, VAP, is required for ER protein quality control. Hum. Mol. Genet., 23, 1975–1989], which mimics many systemic aspects of the human disease. Here, we show that VAPB, located on the cytoplasmic face of the endoplasmic reticulum membrane, interacts with Caspar, an orthologue of human fas associated factor 1 (FAF1). Caspar, in turn, interacts with transitional endoplasmic reticulum ATPase (TER94), a fly orthologue of ALS14 (VCP/p97, valosin-containing protein). Caspar overexpression in the glia extends lifespan and also slows the progression of motor dysfunction in the ALS8 disease model, a phenomenon that we ascribe to its ability to restrain age-dependent inflammation, which is modulated by Relish/NFκB signalling. Caspar binds to VAPB via an FFAT motif, and we find that Caspar’s ability to negatively regulate NFκB signalling is not dependent on the VAPB:Caspar interaction. We hypothesize that Caspar is a key molecule in the pathogenesis of ALS. The VAPB:Caspar:TER94 complex appears to be a candidate for regulating both protein homeostasis and NFκB signalling, with our study highlighting a role for Caspar in glial inflammation. We project human FAF1 as an important protein target to alleviate the progression of motor neuron disease.
Collapse
Affiliation(s)
- Shweta Tendulkar
- Indian Institute of Science Education & Research (IISER) Pune 411008, India
| | - Sushmitha Hegde
- Indian Institute of Science Education & Research (IISER) Pune 411008, India
| | - Lovleen Garg
- Indian Institute of Science Education & Research (IISER) Pune 411008, India
| | | | | | | | | |
Collapse
|
8
|
Kim BS, Song JA, Jang KH, Jang T, Jung B, Yoo SE, Lee JM, Kim E. Pharmacological Intervention Targeting FAF1 Restores Autophagic Flux for α-Synuclein Degradation in the Brain of a Parkinson's Disease Mouse Model. ACS Chem Neurosci 2022; 13:806-817. [PMID: 35230076 DOI: 10.1021/acschemneuro.1c00828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
α-Synuclein accumulation is implicated in the pathogenesis of neurodegenerative diseases, including Parkinson's disease (PD). Previously, we reported that Fas-associated factor 1 (FAF1), which plays a role in PD pathogenesis, potentiates α-synuclein accumulation through autophagy impairment in dopaminergic neurons. In this study, we show that KM-819, a FAF1-targeting compound, which has completed phase I clinical trials, interferes with α-synuclein accumulation in the mouse brain, as well as in human neuronal cells (SH-SY5Ys). KM-819 suppressed the accumulation of monomeric, oligomeric, and aggregated forms of α-synuclein in neuronal cells. Furthermore, KM-819 restored the turnover rate of α-synuclein in FAF1-overexpressing SH-SY5Y cells, implicating KM-819-mediated reconstitution of the α-synuclein degradative pathway. In addition, KM-819 reconstituted autophagic flux in FAF1-transfected SH-SY5Y cells, also suppressing α-synuclein-induced mitochondrial dysfunction. Moreover, oral administration of KM-819 also interfered with α-synuclein accumulation in the midbrain of mice overexpressing FAF1 via an adeno-associated virus system. Consistently, KM-819 reduced α-synuclein accumulation in both the hippocampus and the midbrain of human A53T α-synuclein transgenic mice. Collectively, these data imply that KM-819 may have therapeutic potential for patients with PD.
Collapse
Affiliation(s)
- Bok-Seok Kim
- Department of Biological Sciences, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea
| | - Jin-A Song
- Department of Biological Sciences, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea
| | - Ki-Hong Jang
- Department of Biological Sciences, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea
| | - Taeik Jang
- Department of Biological Sciences, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea
| | - Bumjun Jung
- Department of Biological Sciences, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea
| | | | | | - Eunhee Kim
- Department of Biological Sciences, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Korea
| |
Collapse
|
9
|
Ketkar H, Harrison AG, Graziano VR, Geng T, Yang L, Vella AT, Wang P. UBX Domain Protein 6 Positively Regulates JAK-STAT1/2 Signaling. THE JOURNAL OF IMMUNOLOGY 2021; 206:2682-2691. [PMID: 34021047 DOI: 10.4049/jimmunol.1901337] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 03/24/2021] [Indexed: 01/03/2023]
Abstract
Type I/III IFNs induce expression of hundreds of IFN-stimulated genes through the JAK/STAT pathway to combat viral infections. Although JAK/STAT signaling is seemingly straightforward, it is nevertheless subjected to complex cellular regulation. In this study, we show that an ubiquitination regulatory X (UBX) domain-containing protein, UBXN6, positively regulates JAK-STAT1/2 signaling. Overexpression of UBXN6 enhanced type I/III IFNs-induced expression of IFN-stimulated genes, whereas deletion of UBXN6 inhibited their expression. RNA viral replication was increased in human UBXN6-deficient cells, accompanied by a reduction in both type I/III IFN expression, when compared with UBXN6-sufficient cells. Mechanistically, UBXN6 interacted with tyrosine kinase 2 (TYK2) and inhibited IFN-β-induced degradation of both TYK2 and type I IFNR. These results suggest that UBXN6 maintains normal JAK-STAT1/2 signaling by stabilizing key signaling components during viral infection.
Collapse
Affiliation(s)
- Harshada Ketkar
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT.,Department of Microbiology & Immunology, School of Medicine, New York Medical College, Valhalla, NY; and
| | - Andrew G Harrison
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT
| | - Vincent R Graziano
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT
| | - Tingting Geng
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT
| | - Long Yang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Anthony T Vella
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT
| | - Penghua Wang
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT; .,Department of Microbiology & Immunology, School of Medicine, New York Medical College, Valhalla, NY; and
| |
Collapse
|
10
|
Kim BS, Jang T, Yoo SE, Lee JM, Kim E. Fas-associated factor 1 induces the accumulation of α-synuclein through autophagic suppression in dopaminergic neurons. FASEB J 2021; 35:e21363. [PMID: 33749937 DOI: 10.1096/fj.202001371rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 12/14/2020] [Accepted: 12/28/2020] [Indexed: 12/21/2022]
Abstract
Impairment of protein clearance mechanisms leads to α-synuclein accumulation in dopaminergic neurons, contributing to the pathogenesis of Parkinson's disease (PD). Based on the finding that Fas-associated factor 1 (FAF1), a positive modulator of PD, colocalizes with α-synuclein in PD patient brains, we investigated the existence of pathological interplay between FAF1 and α-synuclein. Monomeric and high-molecular-weight forms of α-synuclein were increased in FAF1-overexpressing SH-SY5Y cells. In particular, α-synuclein turnover was accelerated by genetic depletion of FAF1 in SH-SY5Y cells. Therefore, we questioned whether FAF1 is involved in the α-synuclein clearance process. Autophagy inhibitors, but not proteasome inhibitors, restored concurrent attenuation of α-synuclein expression by FAF1 depletion in SH-SY5Y cells. Moreover, we found alterations in autophagy markers in SH-SY5Y cells caused by FAF1 overexpression, indicating that FAF1 disturbed α-synuclein clearance through the autophagy-lysosome pathway. Indeed, FAF1 activated the mammalian target of rapamycin (mTOR) pathway, subsequently suppressing autophagosome formation. Consistently, α-synuclein-mediated mitochondrial dysfunction was observed in FAF1-overexpressing SH-SY5Y cells. Furthermore, FAF1 overexpression using stereotaxic injection of adeno-associated virus led to α-synuclein accumulation and autophagy dysregulation in the PD model mice. Taken together, our results reveal a novel role for FAF1: that of a negative regulator of autophagic α-synuclein clearance.
Collapse
Affiliation(s)
- Bok-Seok Kim
- Department of Biological Sciences, Chungnam National University, Daejeon, South Korea
| | - Taeik Jang
- Department of Biological Sciences, Chungnam National University, Daejeon, South Korea
| | | | | | - Eunhee Kim
- Department of Biological Sciences, Chungnam National University, Daejeon, South Korea
| |
Collapse
|
11
|
Hu Z, Cao J, Liu G, Zhang H, Liu X. Comparative Transcriptome Profiling of Skeletal Muscle from Black Muscovy Duck at Different Growth Stages Using RNA-seq. Genes (Basel) 2020; 11:genes11101228. [PMID: 33092100 PMCID: PMC7590229 DOI: 10.3390/genes11101228] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/13/2020] [Accepted: 10/16/2020] [Indexed: 12/13/2022] Open
Abstract
In China, the production for duck meat is second only to that of chicken, and the demand for duck meat is also increasing. However, there is still unclear on the internal mechanism of regulating skeletal muscle growth and development in duck. This study aimed to identity candidate genes related to growth of duck skeletal muscle and explore the potential regulatory mechanism. RNA-seq technology was used to compare the transcriptome of skeletal muscles in black Muscovy ducks at different developmental stages (day 17, 21, 27, 31, and 34 of embryos and postnatal 6-month-olds). The SNPs and InDels of black Muscovy ducks at different growth stages were mainly in “INTRON”, “SYNONYMOUS_CODING”, “UTR_3_PRIME”, and “DOWNSTREAM”. The average number of AS in each sample was 37,267, mainly concentrated in TSS and TTS. Besides, a total of 19 to 5377 DEGs were detected in each pairwise comparison. Functional analysis showed that the DEGs were mainly involved in the processes of cell growth, muscle development, and cellular activities (junction, migration, assembly, differentiation, and proliferation). Many of DEGs were well known to be related to growth of skeletal muscle in black Muscovy duck, such as MyoG, FBXO1, MEF2A, and FoxN2. KEGG pathway analysis identified that the DEGs were significantly enriched in the pathways related to the focal adhesion, MAPK signaling pathway and regulation of the actin cytoskeleton. Some DEGs assigned to these pathways were potential candidate genes inducing the difference in muscle growth among the developmental stages, such as FAF1, RGS8, GRB10, SMYD3, and TNNI2. Our study identified several genes and pathways that may participate in the regulation of skeletal muscle growth in black Muscovy duck. These results should serve as an important resource revealing the molecular basis of muscle growth and development in duck.
Collapse
|
12
|
Park G, Kim BS, Kim E. A novel function of FAF1, which induces dopaminergic neuronal death through cell-to-cell transmission. Cell Commun Signal 2020; 18:133. [PMID: 32831099 PMCID: PMC7444258 DOI: 10.1186/s12964-020-00632-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/23/2020] [Indexed: 12/21/2022] Open
Abstract
Background Fas-associated factor 1 (FAF1) has been implicated in Parkinson’s disease (PD) and activates the cell death machinery in the cytosol. However, the presence of extracellular FAF1 has not been studied. Methods Serum-free conditioned medium (CM) from FAF1-transfected SH-SY5Y cells was concentrated and analyzed by western blotting. Exosomes were isolated from CM by ultracentrifugation and analyzed by western blotting, electron microscopy and nanoparticle tracking analysis. Soluble FAF1 from CM was immunodepleted using anti-FAF1 antibody. Transmission of secreted FAF1 was examined by transwell assay under a confocal microscope. CM-induced cell death was determined by measuring propidium iodide (PI) uptake using a flow cytometer. Results FAF1 was secreted from SH-SY5Y cells via exocytosis and brefeldin A (BFA)-resistant secretory pathways. Furthermore, FAF1 was secreted as a vesicle-free form and a genuine exosome cargo in the lumen of exosomes. In addition, FAF1 increased the number of exosomes, suggesting a regulatory role in exosome biogenesis. Extracellular FAF1 was transmitted via endocytosis to neighboring cells, where it induced cell death through apoptotic and necrotic pathways. Conclusions This study presents a novel route by which FAF1 induces neuronal death through cell-to-cell transmission. Video Abstract
Collapse
Affiliation(s)
- Gyeongrin Park
- Department of Biological Sciences, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, South Korea
| | - Bok-Seok Kim
- Department of Biological Sciences, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, South Korea
| | - Eunhee Kim
- Department of Biological Sciences, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, South Korea.
| |
Collapse
|
13
|
Hegde S, Soory A, Kaduskar B, Ratnaparkhi GS. SUMO conjugation regulates immune signalling. Fly (Austin) 2020; 14:62-79. [PMID: 32777975 PMCID: PMC7714519 DOI: 10.1080/19336934.2020.1808402] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/30/2020] [Accepted: 08/05/2020] [Indexed: 12/11/2022] Open
Abstract
Post-translational modifications (PTMs) are critical drivers and attenuators for proteins that regulate immune signalling cascades in host defence. In this review, we explore functional roles for one such PTM, the small ubiquitin-like modifier (SUMO). Very few of the SUMO conjugation targets identified by proteomic studies have been validated in terms of their roles in host defence. Here, we compare and contrast potential SUMO substrate proteins in immune signalling for flies and mammals, with an emphasis on NFκB pathways. We discuss, using the few mechanistic studies that exist for validated targets, the effect of SUMO conjugation on signalling and also explore current molecular models that explain regulation by SUMO. We also discuss in detail roles of evolutionary conservation of mechanisms, SUMO interaction motifs, crosstalk of SUMO with other PTMs, emerging concepts such as group SUMOylation and finally, the potentially transforming roles for genome-editing technologies in studying the effect of PTMs.
Collapse
Affiliation(s)
- Sushmitha Hegde
- Biology, Indian Institute of Science Education & Research (IISER), Pune, India
| | - Amarendranath Soory
- Biology, Indian Institute of Science Education & Research (IISER), Pune, India
| | | | | |
Collapse
|
14
|
Kim TH, Lee HC, Kim JH, Hewawaduge CY, Chathuranga K, Chathuranga WAG, Ekanayaka P, Wijerathne HMSM, Kim CJ, Kim E, Lee JS. Fas-associated factor 1 mediates NADPH oxidase-induced reactive oxygen species production and proinflammatory responses in macrophages against Listeria infection. PLoS Pathog 2019; 15:e1008004. [PMID: 31412082 PMCID: PMC6709923 DOI: 10.1371/journal.ppat.1008004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/26/2019] [Accepted: 07/27/2019] [Indexed: 11/29/2022] Open
Abstract
Fas-associated factor 1 is a death-promoting protein that induces apoptosis by interacting with the Fas receptor. Until now, FAF1 was reported to interact potentially with diverse proteins and to function as a negative and/or positive regulator of several cellular possesses. However, the role of FAF1 in defense against bacterial infection remains unclear. Here, we show that FAF1 plays a pivotal role in activating NADPH oxidase in macrophages during Listeria monocytogenes infection. Upon infection by L. monocytogenes, FAF1 interacts with p67phox (an activator of the NADPH oxidase complex), thereby facilitating its stabilization and increasing the activity of NADPH oxidase. Consequently, knockdown or ectopic expression of FAF1 had a marked effect on production of ROS, proinflammatory cytokines, and antibacterial activity, in macrophages upon stimulation of TLR2 or after infection with L. monocytogenes. Consistent with this, FAF1gt/gt mice, which are knocked down in FAF1, showed weaker inflammatory responses than wild-type mice; these weaker responses led to increased replication of L. monocytogenes. Collectively, these findings suggest that FAF1 positively regulates NADPH oxidase-mediated ROS production and antibacterial defenses. Phagocytic NADPH oxidase plays a pivotal role in generating reactive oxygen species (ROS) and in defense against bacterial infections such as L. monocytogenes. ROS eliminate phagocytosed bacteria directly and are implicated in transduction of signals that mediate inflammatory responses. Here, we show that the apoptotic protein FAF1 regulates ROS production in macrophages by regulating phagocytic NADPH oxidase activity upon infection by L. monocytogenes. FAF1 interacts directly with and stabilizes p67phox, a regulatory protein of the phagocytic NADPH oxidase complex, to induce ROS production during L. monocytogenes infection. Production of ROS leads to release of proinflammatory cytokines, chemokines and, ultimately, to bacterial clearance. Interestingly, FAF1gt/gt mice deficient in FAF1 expression exhibit weakened inflammatory responses and are thus more vulnerable to bacterial infection than FAF1+/+ mice. This study reveals that FAF1 is a crucial regulator that induces inflammatory responses to bacterial infection via ROS production.
Collapse
Affiliation(s)
- Tae-Hwan Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hyun-Cheol Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jae-Hoon Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - C. Y. Hewawaduge
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Kiramage Chathuranga
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | | | - Pathum Ekanayaka
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - H. M. S. M. Wijerathne
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Chul-Joong Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Eunhee Kim
- College of Biological Sciences and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Jong-Soo Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- * E-mail:
| |
Collapse
|
15
|
Identification of two independent SUMO-interacting motifs in Fas-associated factor 1 (FAF1): Implications for mineralocorticoid receptor (MR)-mediated transcriptional regulation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1282-1297. [PMID: 30935967 DOI: 10.1016/j.bbamcr.2019.03.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/19/2019] [Accepted: 03/26/2019] [Indexed: 01/23/2023]
Abstract
Fas-associated factor 1 (FAF1) was originally isolated as a Fas-associated factor and was subsequently found to interact with numerous other proteins that are involved in various cellular events including Fas-mediated apoptosis, nuclear factor (NF)-κB, Wnt/β-catenin, and transforming growth factor (TGF)-β signaling pathways, mineralocorticoid receptor (MR)-mediated transactivation, and ubiquitin-dependent processes. Herein, we defined two small ubiquitin-like modifier (SUMO)-interacting motifs (SIMs) within FAF1 and demonstrated to be crucial for transcriptional modulation of the MR. Our study demonstrated that the SIMs of FAF1 do not play a significant role in regulating its subcellular localization, Fas-mediated apoptosis, or NF-κB or Wnt/β-catenin pathways. Remarkably, FAF1 interacts with the sumoylated MR and represses aldosterone-activated MR transactivation in a SIM-dependent manner. Moreover, silencing of endogenous FAF1 in cells resulted in an increase in the induction of MR target genes by aldosterone, indicating that FAF1 functions as an MR co-repressor. We further provide evidence to suggest that the mechanisms of FAF1/SIM-mediated MR transrepression involve inhibition of MR N/C interactions and promotion of MR polyubiquitination and degradation. Sumoylation has been linked to impacting of repressive properties on several transcription factors and cofactors. Our findings therefore provide mechanistic insights underlying SUMO-dependent transcriptional repression of the MR.
Collapse
|
16
|
Yu C, Kim BS, Park M, Do YJ, Kong YY, Kim E. FAF1 mediates necrosis through JNK1-mediated mitochondrial dysfunction leading to retinal degeneration in the ganglion cell layer upon ischemic insult. Cell Commun Signal 2018; 16:56. [PMID: 30200976 PMCID: PMC6131785 DOI: 10.1186/s12964-018-0265-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/21/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Aberrant cell death induced by ischemic stress is implicated in the pathogenesis of ischemic diseases. Fas-associated factor 1 (FAF1) has been identified as a death-promoting protein. This study demonstrates that FAF1 functions in death signaling triggered by ischemic insult. METHODS The expression changes of FAF1 and phophorylated JNK1 were detected by Western blotting. Immunoprecipitation was employed to investigate protein-protein interaction. We determined the cell death using flow cytometry and lactate dehydrogenase release measurement. To validate the death-promoting role of FAF1 in the retina, we generated conditional retinal FAF1 knockout mice. We used hematoxylin and eosin staining to detect retinal cell death in retinal ganglion cell layer. RESULTS FAF1 was found to function upstream of c-Jun N-terminal kinase 1 (JNK1), followed by mitochondrial dysregulation and necrotic cell death processes upon ischemic insult. We investigated whether FAF1 is involved in the pathogenesis of ischemic diseases using a retinal ischemia model. Indeed, FAF1 potentiated necrosis through JNK1 activation upon ischemic stress in retinal cells demonstrating retinal ganglion-like character. Conditional FAF1 depletion attenuated JNK1 activation in the retinas of Dkk3-Cre;Faf1flox/flox mice and ameliorated death of retinal cells due to elevated intraocular pressure (IOP). CONCLUSIONS Our results show that FAF1 plays a key role in ischemic retinal damage and may be implicated in the pathogenesis of retinal ischemic disease.
Collapse
Affiliation(s)
- Changsun Yu
- Department of Biological Sciences, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134 South Korea
- MOGAM Institute for Biomedical Research, 93, 30beon-gil, Ihyeon-ro, Gilheung-gu, Yongin-si, Gyeonggi-do 16924 South Korea
| | - Bok-seok Kim
- Department of Biological Sciences, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134 South Korea
| | - Minyoung Park
- Department of Biological Sciences, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134 South Korea
- BeyondBio Inc., Daejeon BioVenture Town, 1662, Yuseong-daero, Yuseong-gu, Daejeon, 34134 South Korea
| | - Yun-Ju Do
- Department of Biological Sciences, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134 South Korea
| | - Young-Yun Kong
- School of Biological Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826 South Korea
| | - Eunhee Kim
- Department of Biological Sciences, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134 South Korea
| |
Collapse
|
17
|
Peng H, Huo J, Gao Y, Chen J, Yu X, Xiao T. Fas-associated protein factor 1 is involved in meiotic resumption in mouse oocytes. J Reprod Dev 2018; 64:173-177. [PMID: 29434078 PMCID: PMC5902905 DOI: 10.1262/jrd.2017-081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Fas-associated protein factor 1 (FAF1) is a Fas-associated protein that functions in multiple cellular processes. Previous research showed that mutations in Faf1 led to
the lethality of cleavage stage embryos in a mouse model. The aim of the present study was to analyze the expression pattern, localization, and function of FAF1 in meiotic resumption of
mouse oocytes. FAF1 was exclusively expressed in oocytes at various follicular stages within the ovary and was predominantly localized in the cytoplasm of growing oocytes. Furthermore,
Faf1 mRNA and protein were persistently present during oocyte maturation and Faf1 mRNA levels were similar in the germinal vesicle (GV), GV breakdown
(GVBD), and metaphase II (MII) stages of oocytes. Moreover, knockdown of Faf1 in GV-stage oocytes led to a significantly decreased rate of GVBD. To our knowledge, these
results provide the first evidence regarding a novel function of FAF1 in meiotic resumption in mouse oocytes.
Collapse
Affiliation(s)
- Hui Peng
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
| | - Jianchao Huo
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
| | - Yuyun Gao
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
| | - Jing Chen
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
| | - Xiang Yu
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
| | - Tianfang Xiao
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
| |
Collapse
|
18
|
Liu H, Wang W, Zhang C, Xu C, Duan H, Tian X, Zhang D. Heritability and Genome-Wide Association Study of Plasma Cholesterol in Chinese Adult Twins. Front Endocrinol (Lausanne) 2018; 9:677. [PMID: 30498476 PMCID: PMC6249314 DOI: 10.3389/fendo.2018.00677] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022] Open
Abstract
Dyslipidemia represents a strong and independent risk factor for cardiovascular disease. Plasma cholesterol, such as total cholesterol (TC), low density lipoprotein cholesterol (LDL-C), and high density lipoprotein cholesterol (HDL-C), is the common indicator of diagnosing dyslipidemia. Here based on 382 Chinese twin pairs, we explored the magnitude of genetic impact on TC, HDL-C, LDL-C variation and further searched for genetic susceptibility loci for them using genome-wide association study (GWAS). The ACE model was the best fit model with additive genetic parameter (A) accounting for 26.6%, common or shared environmental parameter (C) accounting for 47.8%, unique/non-shared environmental parameter (E) accounting for 25.6% for the variance in HDL-C. The AE model was the best fit model for TC (A: 61.4%; E: 38.6%) and LDL-C (A: 65.5%; E: 34.5%). While no SNPs reached the genome-wide significance level (P < 5 × 10-8), 8, 14, 9 SNPs exceeded the suggestive significance level (P < 1 × 10-5) for TC, HDL-C, LDL-C, respectively. The promising genetic regions for TC, HDL-C, LDL-C were on chromosome 11 around rs7107698, chromosome 5 around rs12518218, chromosome 2 around rs10490120, respectively. Gene-based analysis found 1038, 1033 and 1090 genes nominally associated with TC, HDL-C, LDL-C (P < 0.05), especially FAF1, KLKB1 for TC, KLKB1 for HDL-C, and NTRK1, FAF1, SNTB2 for LDL-C, respectively. The number of common related genes among TC, HDL-C and LDL-C was 71, including FAF1, KLKB1, etc. Pathway enrichment analysis discovered known related pathways-zinc transporters, metal ion SLC transporters for TC, cell adhesion molecules CAMs, IL-6 signaling for HDL, FC epsilon RI signaling pathway, NFAT pathway for LDL, respectively. In conclusion, the TC and LDL-C level is moderately heritable and the HDL-C level is lowly heritable in Chinese population. The genomic loci, functional genes and pathways are identified to account for the heritability of plasma cholesterol level. Our findings provide important insights into plasma cholesterol molecular physiology and expect future research to replicate and validate our results.
Collapse
Affiliation(s)
- Hui Liu
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Caixia Zhang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Chunsheng Xu
- Qingdao Municipal Centre for Disease Control and Prevention, Qingdao, China
| | - Haiping Duan
- Qingdao Municipal Centre for Disease Control and Prevention, Qingdao, China
| | - Xiaocao Tian
- Qingdao Municipal Centre for Disease Control and Prevention, Qingdao, China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
- *Correspondence: Dongfeng Zhang
| |
Collapse
|
19
|
Kim JH, Park ME, Nikapitiya C, Kim TH, Uddin MB, Lee HC, Kim E, Ma JY, Jung JU, Kim CJ, Lee JS. FAS-associated factor-1 positively regulates type I interferon response to RNA virus infection by targeting NLRX1. PLoS Pathog 2017; 13:e1006398. [PMID: 28542569 PMCID: PMC5456407 DOI: 10.1371/journal.ppat.1006398] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 06/02/2017] [Accepted: 05/04/2017] [Indexed: 12/25/2022] Open
Abstract
FAS-associated factor-1 (FAF1) is a component of the death-inducing signaling complex involved in Fas-mediated apoptosis. It regulates NF-κB activity, ubiquitination, and proteasomal degradation. Here, we found that FAF1 positively regulates the type I interferon pathway. FAF1gt/gt mice, which deficient in FAF1, and FAF1 knockdown immune cells were highly susceptible to RNA virus infection and showed low levels of inflammatory cytokines and type I interferon (IFN) production. FAF1 was bound competitively to NLRX1 and positively regulated type I IFN signaling by interfering with the interaction between NLRX1 and MAVS, thereby freeing MAVS to bind RIG-I, which switched on the MAVS-RIG-I-mediated antiviral signaling cascade. These results highlight a critical role of FAF1 in antiviral responses against RNA virus infection.
Collapse
Affiliation(s)
- Jae-Hoon Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Min-Eun Park
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Chamilani Nikapitiya
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Tae-Hwan Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Md Bashir Uddin
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- Faculty of Veterinary & Animal Science, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Hyun-Cheol Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Eunhee Kim
- College of Biological Sciences and Biotechnology, Chungnam National University, Daejeon, Korea
| | - Jin Yeul Ma
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu, Republic of Korea
| | - Jae U. Jung
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, California, United States of America
| | - Chul-Joong Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jong-Soo Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
20
|
XIAP Interacts with and Regulates the Activity of FAF1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1335-1348. [PMID: 28414080 DOI: 10.1016/j.bbamcr.2017.04.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 03/26/2017] [Accepted: 04/12/2017] [Indexed: 12/19/2022]
Abstract
Cell death depends on the balance between the activities of pro- and anti-apoptotic factors. X-linked inhibitor of apoptosis protein (XIAP) plays an important role in the cytoprotective process by inhibiting the caspase cascade and regulating pro-survival signaling pathways. While searching for novel interacting partners of XIAP, we identified Fas-associated factor 1 (FAF1). Contrary to XIAP, FAF1 is a pro-apoptotic factor that also regulates several signaling pathways in which XIAP is involved. However, the functional relationship between FAF1 and XIAP is unknown. Here, we describe a new interaction between XIAP and FAF1 and describe the functional implications of their opposing roles in cell death and NF-κB signaling. Our results clearly demonstrate the interaction of XIAP with FAF1 and define the specific region of the interaction. We observed that XIAP is able to block FAF1-mediated cell death by interfering with the caspase cascade and directly interferes in NF-κB pathway inhibition by FAF1. Furthermore, we show that XIAP promotes ubiquitination of FAF1. Conversely, FAF1 does not interfere with the anti-apoptotic activity of XIAP, despite binding to the BIR domains of XIAP; however, FAF1 does attenuate XIAP-mediated NF-κB activation. Altered expression of both factors has been implicated in degenerative and cancerous processes; therefore, studying the balance between XIAP and FAF1 in these pathologies will aid in the development of novel therapies.
Collapse
|
21
|
Yu C, Kim BS, Kim E. FAF1 mediates regulated necrosis through PARP1 activation upon oxidative stress leading to dopaminergic neurodegeneration. Cell Death Differ 2016; 23:1873-1885. [PMID: 27662363 PMCID: PMC5071579 DOI: 10.1038/cdd.2016.99] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 07/29/2016] [Accepted: 08/17/2016] [Indexed: 02/02/2023] Open
Abstract
Cumulative damage caused by oxidative stress results in diverse pathological conditions. Therefore, elucidating the molecular mechanisms underlying cell death following oxidative stress is important. Here, we describe a novel role for Fas-associated factor 1 (FAF1) as a crucial regulator of necrotic cell death elicited by hydrogen peroxide. Upon oxidative insult, FAF1 translocated from the cytoplasm to the nucleus and promoted the catalytic activation of poly(ADP-ribose) polymerase 1 (PARP1) through physical interaction. Moreover, FAF1 depletion prevented PARP1-linked downstream events involved in the triggering of cell death, including energetic collapse, mitochondrial depolarization and nuclear translocation of apoptosis-inducing factor (AIF), implying that FAF1 has a key role in PARP1-dependent necrosis in response to oxidative stress. We further investigated whether FAF1 might contribute to the pathogenesis of Parkinson's disease through excessive PARP1 activation. Indeed, the overexpression of FAF1 using a recombinant adeno-associated virus system in the mouse ventral midbrain promoted PARP1 activation and dopaminergic neurodegeneration in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease. Collectively, our data demonstrate the presence of an FAF1-PARP1 axis that is involved in oxidative stress-induced necrosis and in the pathology of Parkinson's disease.
Collapse
Affiliation(s)
- Changsun Yu
- Department of Biological Sciences, Chungnam National University, Daejeon 305-764, Korea
| | - Bok-seok Kim
- Department of Biological Sciences, Chungnam National University, Daejeon 305-764, Korea
| | - Eunhee Kim
- Department of Biological Sciences, Chungnam National University, Daejeon 305-764, Korea
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 305-764, Korea
| |
Collapse
|
22
|
Rezvani K. UBXD Proteins: A Family of Proteins with Diverse Functions in Cancer. Int J Mol Sci 2016; 17:ijms17101724. [PMID: 27754413 PMCID: PMC5085755 DOI: 10.3390/ijms17101724] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 09/24/2016] [Accepted: 10/08/2016] [Indexed: 12/15/2022] Open
Abstract
The UBXD family is a diverse group of UBX (ubiquitin-regulatory X) domain-containing proteins in mammalian cells. Members of this family contain a UBX domain typically located at the carboxyl-terminal of the protein. In contrast to the UBX domain shared by all members of UBXD family, the amino-terminal domains are diverse and appear to carry out different roles in a subcellular localization-dependent manner. UBXD proteins are principally associated with the endoplasmic reticulum (ER), where they positively or negatively regulate the ER-associated degradation machinery (ERAD). The distinct protein interaction networks of UBXD proteins allow them to have specific functions independent of the ERAD pathway in a cell type- and tissue context-dependent manner. Recent reports have illustrated that a number of mammalian members of the UBXD family play critical roles in several proliferation and apoptosis pathways dysregulated in selected types of cancer. This review covers recent advances that elucidate the therapeutic potential of selected members of the UBXD family that can contribute to tumor growth.
Collapse
Affiliation(s)
- Khosrow Rezvani
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD 57069, USA.
| |
Collapse
|
23
|
Rao P, Zhou Y, Ge SQ, Wang AX, Yu XW, Alzain MA, Veronica AK, Qiu J, Song MS, Zhang J, Wang H, Fang HH, Gao Q, Wang YX, Wang W. Validation of Type 2 Diabetes Risk Variants Identified by Genome-Wide Association Studies in Northern Han Chinese. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:ijerph13090863. [PMID: 27589775 PMCID: PMC5036696 DOI: 10.3390/ijerph13090863] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/05/2016] [Accepted: 08/23/2016] [Indexed: 01/07/2023]
Abstract
Background: More than 60 genetic susceptibility loci associated with type 2 diabetes mellitus (T2DM) have been established in populations of Asian and European ancestry. Given ethnic differences and environmental factors, validation of the effects of genetic risk variants with reported associations identified by Genome-Wide Association Studies (GWASs) is essential. The study aims at evaluating the associations of T2DM with 29 single nucleotide polymorphisms (SNPs) from 19 candidate genes derived from GWASs in a northern Han Chinese population. Method: In this case-control study, 461 T2DM-diagnosed patients and 434 controls were recruited at the Jidong oil field hospital (Hebei, China) from January 2009 to October 2013. A cumulative genetic risk score (cGRS) was calculated by summation of the number of risk alleles, and a weight GRS (wGRS) was calculated as the sum of risk alleles at each locus multiplied by their effect sizes for T2DM, using the independent variants selected. Result: The allelic frequency of the “A” allele at rs17106184 (Fas-associated factor 1, FAF1) was significantly higher in the T2DM patients than that of the healthy controls (11.7% vs. 6.4%, p < 0.001). Individuals in the highestquartile of wGRS had an over three-fold increased risk for developing T2DM compared with those in the lowest quartile (odds ratio = 3.06, 95% CI = 1.92–4.88, p < 0.001) adjusted for age, sex, BMI, total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), systolic blood pressure (SBP) and diastolic blood pressure (DBP). The results were similar when analyzed with the cGRS. Conclusions: We confirmed the association between rs17106184 (FAF1) and T2DM in a northern Han Chinese population. The GRS calculated based on T2DM susceptibility variants may be a useful tool for predicting the T2DM susceptibility.
Collapse
Affiliation(s)
- Ping Rao
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China.
- Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China.
| | - Yong Zhou
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| | - Si-Qi Ge
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China.
- School of Medical Sciences, Edith Cowan University, Perth, WA 6027, Australia.
| | - An-Xin Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China.
| | - Xin-Wei Yu
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China.
- School of Medical Sciences, Edith Cowan University, Perth, WA 6027, Australia.
| | - Mohamed Ali Alzain
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China.
| | - Andrea Katherine Veronica
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China.
| | - Jing Qiu
- School of Public Health, Ningxia Medical University, Yinchuan 750021, China.
| | - Man-Shu Song
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China.
| | - Jie Zhang
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China.
| | - Hao Wang
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China.
| | - Hong-Hong Fang
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China.
| | - Qing Gao
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China.
| | - You-Xin Wang
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China.
| | - Wei Wang
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing 100069, China.
- School of Medical Sciences, Edith Cowan University, Perth, WA 6027, Australia.
| |
Collapse
|
24
|
Ma X, Li X, Dong S, Xia Q, Wang F. A Fas associated factor negatively regulates anti-bacterial immunity by promoting Relish degradation in Bombyx mori. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2015; 63:144-151. [PMID: 26101847 DOI: 10.1016/j.ibmb.2015.06.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/15/2015] [Accepted: 06/18/2015] [Indexed: 06/04/2023]
Abstract
Negative regulation is required to keep NF-κB-dependent immune response under tight control. In previous study, we have identified a Fas associated factor (FAF) family member in Bombyx mori, BmFAF, and proposed it may act as a negative regulator in immune response. In this study, we found knock-down of BmFAF by RNAi led to a remarkable increase in transcriptional level of several antimicrobial peptide genes, including BmCecropinA1 and BmMoricin, and higher survival rate to Gram-negative bacterial infection. We also confirmed the regulatory role of BmFAF in suppressing NF-κB-dependent transcription by employing an inducible promoter in BmE cells. Consistent with these physiological phenotypes, BmFAF suppressed the activity of the essential transcription factor, Relish, in IMD signaling pathway by promoting its proteasomal degradation through direct interaction. In addition, by constructing various truncation mutants, we further demonstrated that UBA domain in BmFAF is required for the inhibitory role, and potential ubiquitination also occurs in this domain. Taken together, our results suggest that BmFAF is a negative regulator of IMD pathway by mediating degradation of Relish.
Collapse
Affiliation(s)
- Xiaojuan Ma
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China
| | - Xianyang Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China
| | - Shifeng Dong
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China
| | - Qingyou Xia
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China
| | - Fei Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China.
| |
Collapse
|
25
|
Wang YB, Tan B, Mu R, Chang Y, Wu M, Tu HQ, Zhang YC, Guo SS, Qin XH, Li T, Li WH, Li AL, Zhang XM, Li HY. Ubiquitin-associated domain-containing ubiquitin regulatory X (UBX) protein UBXN1 is a negative regulator of nuclear factor κB (NF-κB) signaling. J Biol Chem 2015; 290:10395-405. [PMID: 25681446 DOI: 10.1074/jbc.m114.631689] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Indexed: 01/17/2023] Open
Abstract
Excessive nuclear factor κB (NF-κB) activation should be precisely controlled as it contributes to multiple immune and inflammatory diseases. However, the negative regulatory mechanisms of NF-κB activation still need to be elucidated. Various types of polyubiquitin chains have proved to be involved in the process of NF-κB activation. Many negative regulators linked to ubiquitination, such as A20 and CYLD, inhibit IκB kinase activation in the NF-κB signaling pathway. To find new NF-κB signaling regulators linked to ubiquitination, we used a small scale siRNA library against 51 ubiquitin-associated domain-containing proteins and screened out UBXN1, which contained both ubiquitin-associated and ubiquitin regulatory X (UBX) domains as a negative regulator of TNFα-triggered NF-κB activation. Overexpression of UBXN1 inhibited TNFα-triggered NF-κB activation, although knockdown of UBXN1 had the opposite effect. UBX domain-containing proteins usually act as valosin-containing protein (VCP)/p97 cofactors. However, knockdown of VCP/p97 barely affected UBXN1-mediated NF-κB inhibition. At the same time, we found that UBXN1 interacted with cellular inhibitors of apoptosis proteins (cIAPs), E3 ubiquitin ligases of RIP1 in the TNFα receptor complex. UBXN1 competitively bound to cIAP1, blocked cIAP1 recruitment to TNFR1, and sequentially inhibited RIP1 polyubiquitination in response to TNFα. Therefore, our findings demonstrate that UBXN1 is an important negative regulator of the TNFα-triggered NF-κB signaling pathway by mediating cIAP recruitment independent of VCP/p97.
Collapse
Affiliation(s)
- Yu-Bo Wang
- From the Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Bo Tan
- From the Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Rui Mu
- From the Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Yan Chang
- From the Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Min Wu
- From the Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Hai-Qing Tu
- From the Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Yu-Cheng Zhang
- From the Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Sai-Sai Guo
- From the Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Xuan-He Qin
- From the Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Tao Li
- From the Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Wei-Hua Li
- From the Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Ai-Ling Li
- From the Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Xue-Min Zhang
- From the Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| | - Hui-Yan Li
- From the Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing 100850, China
| |
Collapse
|
26
|
Baron Y, Pedrioli PG, Tyagi K, Johnson C, Wood NT, Fountaine D, Wightman M, Alexandru G. VAPB/ALS8 interacts with FFAT-like proteins including the p97 cofactor FAF1 and the ASNA1 ATPase. BMC Biol 2014; 12:39. [PMID: 24885147 PMCID: PMC4068158 DOI: 10.1186/1741-7007-12-39] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 05/12/2014] [Indexed: 11/10/2022] Open
Abstract
Background FAF1 is a ubiquitin-binding adaptor for the p97 ATPase and belongs to the UBA-UBX family of p97 cofactors. p97 converts the energy derived from ATP hydrolysis into conformational changes of the p97 hexamer, which allows the dissociation of its targets from cellular structures or from larger protein complexes to facilitate their ubiquitin-dependent degradation. VAPB and the related protein VAPA form homo- and heterodimers that are anchored in the endoplasmic reticulum membrane and can interact with protein partners carrying a FFAT motif. Mutations in either VAPB or p97 can cause amyotrophic lateral sclerosis, a neurodegenerative disorder that affects upper and lower motor neurons. Results We show that FAF1 contains a non-canonical FFAT motif that allows it to interact directly with the MSP domain of VAPB and, thereby, to mediate VAPB interaction with p97. This finding establishes a link between two proteins that can cause amyotrophic lateral sclerosis when mutated, VAPB/ALS8 and p97/ALS14. Subsequently, we identified a similar FFAT-like motif in the ASNA1 subunit of the transmembrane-domain recognition complex (TRC), which in turn mediates ASNA1 interaction with the MSP domain of VAPB. Proteasome inhibition leads to the accumulation of ubiquitinated species in VAPB immunoprecipitates and this correlates with an increase in FAF1 and p97 binding. We found that VAPB interaction with ubiquitinated proteins is strongly reduced in cells treated with FAF1 siRNA. Our efforts to determine the identity of the ubiquitinated targets common to VAPB and FAF1 led to the identification of RPN2, a subunit of an oligosaccharyl-transferase located at the endoplasmic reticulum, which may be regulated by ubiquitin-mediated degradation. Conclusions The FFAT-like motifs we identified in FAF1 and ASNA1 demonstrate that sequences containing a single phenylalanine residue with the consensus (D/E)(D/E)FEDAx(D/E) are also proficient to mediate interaction with VAPB. Our findings indicate that the repertoire of VAPB interactors is more diverse than previously anticipated and link VAPB to the function of ATPase complexes such as p97/FAF1 and ASNA1/TRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Gabriela Alexandru
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit (MRC-PPU), College of Life Sciences, University of Dundee, Dow St, Dundee DD1 5EH, UK.
| |
Collapse
|
27
|
Nagashima R, Kawakami F, Takahashi S, Obata F, Kubo M. Allo-antigen stimulated CD8+ T-cells suppress NF-κB and Ets-1 DNA binding activity, and inhibit phosphorylated NF-κB p65 nuclear localization in CD4+ T-cells. Viral Immunol 2014; 27:305-15. [PMID: 24844121 DOI: 10.1089/vim.2013.0113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
CD8+ T-cells of asymptomatic HIV-1 carriers (AC) suppress human immunodeficiency virus type 1 (HIV-1) replication in a class I major histocompatibility complex (MHC-I)-restricted and -unrestricted manner. In order to investigate the mechanism of MHC-I-unrestricted CD8+ T-cell-mediated HIV-1 suppression, we previously established allo-antigen stimulated CD8+T-cells from HIV-1-uninfected donors. These allo-antigen stimulated CD8+ T-cells suppressed HIV-1 replication in acutely infected autologous CD4+ T-cells when directly co-cultured. To elucidate the mechanism of HIV-1 replication suppression, we analyzed DNA-binding activity and phosphorylation of transcriptional factors associated with HIV-1 replication by electrophoresis mobility shift assay and Western blotting. When CD4+ T-cells were cultured with allo-antigen stimulated CD8+ T-cells, the reduction of NF-κB and Ets-1 DNA-binding activity was observed. Nuclear localization of NF-κB p65 and Ets-1 was suppressed in CD4+ T-cells. Although NF-κB p65 and Ets-1 are known to be regulated by protein kinase A (PKA), no difference was observed in the expression and phosphorylation of the PKA catalytic subunit in CD4+ T-cells cultured with PHA-treated CD8+ T-cells or allo-antigen stimulated CD8+ T-cells. Cyclic AMP is also known to enter through gap junctions, but the suppression of HIV-1 replication mediated by allo-antigen stimulated CD8+ T-cells was not affected by the gap junction inhibitor. The nuclear transport of phosphorylated NF-κB p65 (Ser276) was inhibited only in CD4+ T-cells cultured with allo-antigen stimulated CD8+ T-cells. Our results indicate that allo-antigen stimulated CD8+ T-cells suppress the transcriptional activity of NF-κB p65 or Ets-1 in an antigen-nonspecific manner, and inhibit the nuclear transport of phosphorylated NF-κB p65 (Ser276).
Collapse
Affiliation(s)
- Ryuichi Nagashima
- 1 Division of Clinical Immunology, Graduate School of Medical Sciences, Kitasato University , Sagamihara, Japan
| | | | | | | | | |
Collapse
|
28
|
Fas-associated factor (Faf1) is a novel CD40 interactor that regulates CD40-induced NF-κB activation via a negative feedback loop. Cell Death Dis 2014; 5:e1213. [PMID: 24810049 PMCID: PMC4047894 DOI: 10.1038/cddis.2014.172] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 02/03/2014] [Accepted: 02/13/2014] [Indexed: 12/24/2022]
Abstract
CD40-induced signalling through ligation with its natural ligand (CD40L/CD154) is dependent on recruitment of TRAF molecules to the cytoplasmic domain of the receptor. Here, we applied the yeast two-hybrid system to examine whether other proteins can interact with CD40. Fas-Associated Factor 1(FAF1) was isolated from a HeLa cDNA library using the CD40 cytoplasmic tail (216–278 aa) as a bait construct. FAF1 was able to interact with CD40 both in vitro and in vivo. The FAF1 N-terminal domain was sufficient to bind CD40 and required the TRAF6-binding domain within the cytoplasmic tail of CD40 for binding. CD40 ligation induced FAF1 expression in an NFκB-dependent manner. Knockdown of FAF1 prolonged CD40-induced NFκB, whereas overexpression of FAF1 suppressed CD40-induced NFκB activity and this required interaction of FAF1 with the CD40 receptor via its FID domain. Thus, we report a novel role for FAF1in regulating CD40-induced NFκB activation via a negative feedback loop. Loss of FAF1 function in certain human malignancies may contribute to oncogenesis through unchecked NFκB activation, and further understanding of this process may provide a biomarker of NFκB-targeted therapies for such malignancies.
Collapse
|
29
|
Kleino A, Silverman N. The Drosophila IMD pathway in the activation of the humoral immune response. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 42:25-35. [PMID: 23721820 PMCID: PMC3808521 DOI: 10.1016/j.dci.2013.05.014] [Citation(s) in RCA: 236] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Revised: 05/17/2013] [Accepted: 05/17/2013] [Indexed: 05/08/2023]
Abstract
The IMD pathway signaling plays a pivotal role in the Drosophila defense against bacteria. During the last two decades, significant progress has been made in identifying the components and deciphering the molecular mechanisms underlying this pathway, including the means of bacterial sensing and signal transduction. While these findings have contributed to the understanding of the immune signaling in insects, they have also provided new insights in studying the mammalian NF-κB signaling pathways. Here, we summarize the current view of the IMD pathway focusing on how it regulates the humoral immune response of Drosophila.
Collapse
Affiliation(s)
- Anni Kleino
- Division of Infectious Diseases, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | | |
Collapse
|
30
|
The p97-UFD1L-NPL4 protein complex mediates cytokine-induced IκBα proteolysis. Mol Cell Biol 2013; 34:335-47. [PMID: 24248593 DOI: 10.1128/mcb.01190-13] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
IκBα is an inhibitor of NF-κB, a family of transcription factors that transactivate genes related to inflammation. Upon inflammatory stimuli, IκBα is rapidly degraded via the ubiquitin-proteasome pathway. While it is very clear that the SCF(β-TRCP) ubiquitin ligase ubiquitinates IκBα upon stimulation, little is known about the postubiquitinational events of IκBα proteolysis. Here, we report that p97, a valosin-containing protein (also called VCP), plays an essential role in the postubiquitinational regulation of IκBα turnover after tumor necrosis factor alpha (TNF-α) or interleukin-1β (IL-1β) treatment. The ATPase activity of p97 is essential for its role in IκBα proteolysis. Moreover, we found that UFD1L and NPL4, two cofactors of p97, assist p97 to control the postubiquitinational regulation of IκBα. The p97-UFD1L-NPL4 protein complex specifically associates with ubiquitinated IκBα via the interactions between p97 and the SCF(β-TRCP) ubiquitin ligase and between the polyubiquitin binding domain of UFD1L and polyubiquitinated IκBα. Furthermore, we observed that the postubiquitinational regulation of IκBα by the p97-UFD1L-NPL4 complex is important for NF-κB activation under stimuli.
Collapse
|
31
|
Kim H, Zhang H, Meng D, Russell G, Lee JN, Ye J. UAS domain of Ubxd8 and FAF1 polymerizes upon interaction with long-chain unsaturated fatty acids. J Lipid Res 2013; 54:2144-2152. [PMID: 23720822 DOI: 10.1194/jlr.m037218] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Ubxd8, a multidomain protein sensor for long-chain unsaturated fatty acids (FAs), plays a crucial role to maintain cellular homeostasis of FAs. Ubxd8 polymerizes upon interaction with long-chain unsaturated FAs, but the molecular mechanism involved in this polymerization remains unclear. Here we report that the UAS domain of Ubxd8 mediates this polymerization. We show that a positively charged surface area in the domain is required for the reaction. Mutations changing the positively charged residues in this area to glutamates prevented long-chain unsaturated FAs from inducing oligomerization of Ubxd8. Consequently, the mutant protein no longer responded to regulation by long-chain unsaturated FAs in cultured cells. Long-chain unsaturated FAs also induced polymerization of Fas-associated factor 1 (FAF1), the only other mammalian protein that contains a UAS domain homologous to that of Ubxd8. These results provide further insights into protein-FA interactions by identifying the UAS domain as a motif interacting with long-chain unsaturated FAs.
Collapse
Affiliation(s)
- Hyeonwoo Kim
- Departments of Molecular Genetics University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Hong Zhang
- Biophysics University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - David Meng
- Departments of Molecular Genetics University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Geoffrey Russell
- Departments of Molecular Genetics University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Joon No Lee
- Departments of Molecular Genetics University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jin Ye
- Departments of Molecular Genetics University of Texas Southwestern Medical Center, Dallas, TX 75390.
| |
Collapse
|
32
|
Franz A, Ackermann L, Hoppe T. Create and preserve: proteostasis in development and aging is governed by Cdc48/p97/VCP. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1843:205-15. [PMID: 23583830 DOI: 10.1016/j.bbamcr.2013.03.031] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 03/08/2013] [Accepted: 03/25/2013] [Indexed: 12/24/2022]
Abstract
The AAA-ATPase Cdc48 (also called p97 or VCP) acts as a key regulator in proteolytic pathways, coordinating recruitment and targeting of substrate proteins to the 26S proteasome or lysosomal degradation. However, in contrast to the well-known function in ubiquitin-dependent cellular processes, the physiological relevance of Cdc48 in organismic development and maintenance of protein homeostasis is less understood. Therefore, studies on multicellular model organisms help to decipher how Cdc48-dependent proteolysis is regulated in time and space to meet developmental requirements. Given the importance of developmental regulation and tissue maintenance, defects in Cdc48 activity have been linked to several human pathologies including protein aggregation diseases. Thus, addressing the underlying disease mechanisms not only contributes to our understanding on the organism-wide function of Cdc48 but also facilitates the design of specific medical therapies. In this review, we will portray the role of Cdc48 in the context of multicellular organisms, pointing out its importance for developmental processes, tissue surveillance, and disease prevention. This article is part of a Special Issue entitled: Ubiquitin-Proteasome System. Guest Editors: Thomas Sommer and Dieter H. Wolf.
Collapse
Affiliation(s)
- André Franz
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Zülpicher Straße 47a, 50674 Cologne, Germany
| | | | | |
Collapse
|
33
|
Lee JJ, Park JK, Jeong J, Jeon H, Yoon JB, Kim EE, Lee KJ. Complex of Fas-associated factor 1 (FAF1) with valosin-containing protein (VCP)-Npl4-Ufd1 and polyubiquitinated proteins promotes endoplasmic reticulum-associated degradation (ERAD). J Biol Chem 2013; 288:6998-7011. [PMID: 23293021 DOI: 10.1074/jbc.m112.417576] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Fas-associated factor 1 (FAF1) is a ubiquitin receptor containing multiple ubiquitin-related domains including ubiquitin-associated (UBA), ubiquitin-like (UBL) 1, UBL2, and ubiquitin regulatory X (UBX). We previously showed that N-terminal UBA domain recognizes Lys(48)-ubiquitin linkage to recruit polyubiquitinated proteins and that a C-terminal UBX domain interacts with valosin-containing protein (VCP). This study shows that FAF1 interacts only with VCP complexed with Npl4-Ufd1 heterodimer, a requirement for the recruitment of polyubiquitinated proteins to UBA domain. Intriguingly, VCP association to C-terminal UBX domain regulates ubiquitin binding to N-terminal UBA domain without direct interaction between UBA and UBX domains. These interactions are well characterized by structural and biochemical analysis. VCP-Npl4-Ufd1 complex is known as the machinery required for endoplasmic reticulum-associated degradation. We demonstrate here that FAF1 binds to VCP-Npl4-Ufd1 complex via UBX domain and polyubiquitinated proteins via UBA domain to promote endoplasmic reticulum-associated degradation.
Collapse
Affiliation(s)
- Jae-Jin Lee
- Center for Cell Signaling and Drug Discovery Research, College of Pharmacy, Division of Life and Pharmaceutical Sciences, Department of Bioinspired Science, Ewha Womans University, Seoul 120-750, Korea
| | | | | | | | | | | | | |
Collapse
|
34
|
Lee JJ, Kim YM, Jeong J, Bae DS, Lee KJ. Ubiquitin-associated (UBA) domain in human Fas associated factor 1 inhibits tumor formation by promoting Hsp70 degradation. PLoS One 2012; 7:e40361. [PMID: 22876279 PMCID: PMC3410879 DOI: 10.1371/journal.pone.0040361] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 06/06/2012] [Indexed: 02/02/2023] Open
Abstract
Human Fas associated factor 1 (hFAF1) is a pro-apoptotic scaffolding protein containing ubiquitin-associating (UBA), ubiquitin like 1 and 2 (UBL1, UBL2), and ubiquitin regulatory X (UBX) domains. hFAF1 interacts with polyubiquitinated proteins via its N-terminal UBA domain and with valosin containing protein (VCP) via its C-terminal UBX domain. Overexpression of hFAF1 or its N-terminal UBA domain significantly increases cell death by increasing the degradation of polyubiquitinated proteins. In this study, we investigated whether hFAF1, whose expression level is reduced in cervical cancer, plays a role in tumor formation. We found that HeLa cells overexpressing full-length hFAF1 or the hFAF1 UBA domain alone, significantly suppressed the anchorage independent tumor growth in soft agar colony formation, increased cell death, and activated JNK and caspase 3. Employing UBA-specific tandem immunoprecipitation, we identified moieties specifically interacting with UBA domain of hFAF1, and found that polyubiquitinated Hsp70s are recruited to UBA domain. We also demonstrated that hFAF1 overexpression promotes Hsp70 degradation via the proteasome. We further found that mutating the UBA domain (I41N), as well as knocking down hFAF1 with specific RNAi, abolishs its ability to increase the proteasomal degradation of Hsp70. These findings suggest that hFAF1 inhibits tumor formation by increasing the degradation of Hsp70 mediated via its UBA domain.
Collapse
Affiliation(s)
- Jae-Jin Lee
- Division of Life and Pharmaceutical Sciences, Department of Bioinspired Science, Drug Discovery Research, College of Pharmacy, Center for Cell Signaling, Ewha Womans University, Seoul, Korea
| | - Young Mee Kim
- Division of Life and Pharmaceutical Sciences, Department of Bioinspired Science, Drug Discovery Research, College of Pharmacy, Center for Cell Signaling, Ewha Womans University, Seoul, Korea
| | - Jaeho Jeong
- Division of Life and Pharmaceutical Sciences, Department of Bioinspired Science, Drug Discovery Research, College of Pharmacy, Center for Cell Signaling, Ewha Womans University, Seoul, Korea
| | - Duk Soo Bae
- Department of Obstetrics and Gynecology, Samsung Medical Center, SungKyunKwan University, Seoul, Korea
| | - Kong-Joo Lee
- Division of Life and Pharmaceutical Sciences, Department of Bioinspired Science, Drug Discovery Research, College of Pharmacy, Center for Cell Signaling, Ewha Womans University, Seoul, Korea
- * E-mail:
| |
Collapse
|
35
|
Zhang L, Zhou F, Li Y, Drabsch Y, Zhang J, van Dam H, ten Dijke P. Fas-associated factor 1 is a scaffold protein that promotes β-transducin repeat-containing protein (β-TrCP)-mediated β-catenin ubiquitination and degradation. J Biol Chem 2012; 287:30701-10. [PMID: 22730322 DOI: 10.1074/jbc.m112.353524] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
FAS-associated factor 1 (FAF1) antagonizes Wnt signaling by stimulating β-catenin degradation. However, the molecular mechanism underlying this effect is unknown. Here, we demonstrate that the E3 ubiquitin ligase β-transducin repeat-containing protein (β-TrCP) is required for FAF1 to suppress Wnt signaling and that FAF1 specifically associates with the SCF (Skp1-Cul1-F-box protein)-β-TrCP complex. Depletion of β-TrCP reduced FAF1-mediated β-catenin polyubiquitination and impaired FAF1 in antagonizing Wnt/β-catenin signaling. FAF1 was shown to act as a scaffold for β-catenin and β-TrCP and thereby to potentiate β-TrCP-mediated β-catenin ubiquitination and degradation. Data mining revealed that FAF1 expression is statistically down-regulated in human breast carcinoma compared with normal breast tissue. Consistent with this, FAF1 expression is higher in epithelial-like MCF7 than mesenchymal-like MDA-MB-231 human breast cancer cells. Depletion of FAF1 in MCF7 cells resulted in increased β-catenin accumulation and signaling. Importantly, FAF1 knockdown promoted a decrease in epithelial E-cadherin and an increase in mesenchymal vimentin expression, indicative for an epithelial to mesenchymal transition. Moreover, ectopic FAF1 expression reduces breast cancer cell migration in vitro and invasion/metastasis in vivo. Thus, our studies strengthen a tumor-suppressive function for FAF1.
Collapse
Affiliation(s)
- Long Zhang
- Department of Molecular Cell Biology and Centre for Biomedical Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
36
|
Pinheiro AS, Eibl C, Ekman-Vural Z, Schwarzenbacher R, Peti W. The NLRP12 pyrin domain: structure, dynamics, and functional insights. J Mol Biol 2011; 413:790-803. [PMID: 21978668 DOI: 10.1016/j.jmb.2011.09.024] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 09/12/2011] [Accepted: 09/14/2011] [Indexed: 01/12/2023]
Abstract
The initial line of defense against infection is sustained by the innate immune system. Together, membrane-bound Toll-like receptors and cytosolic nucleotide-binding domain and leucine-rich repeat-containing receptors (NLR) play key roles in the innate immune response by detecting bacterial and viral invaders as well as endogenous stress signals. NLRs are multi-domain proteins with varying N-terminal effector domains that are responsible for regulating downstream signaling events. Here, we report the structure and dynamics of the N-terminal pyrin domain of NLRP12 (NLRP12 PYD) determined using NMR spectroscopy. NLRP12 is a non-inflammasome NLR that has been implicated in the regulation of Toll-like receptor-dependent nuclear factor-κB activation. NLRP12 PYD adopts a typical six-helical bundle death domain fold. By direct comparison with other PYD structures, we identified hydrophobic residues that are essential for the stable fold of the NLRP PYD family. In addition, we report the first in vitro confirmed non-homotypic PYD interaction between NLRP12 PYD and the pro-apoptotic protein Fas-associated factor 1 (FAF-1), which links the innate immune system to apoptotic signaling. Interestingly, all residues that participate in this protein:protein interaction are confined to the α2-α3 surface, a region of NLRP12 PYD that differs most between currently reported NLRP PYD structures. Finally, we experimentally highlight a significant role for tryptophan 45 in the interaction between NLRP12 PYD and the FAF-1 UBA domain.
Collapse
Affiliation(s)
- Anderson S Pinheiro
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, RI 02903, USA
| | | | | | | | | |
Collapse
|
37
|
Crystal structure of human FAF1 UBX domain reveals a novel FcisP touch-turn motif in p97/VCP-binding region. Biochem Biophys Res Commun 2011; 407:531-4. [PMID: 21414298 DOI: 10.1016/j.bbrc.2011.03.052] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 03/11/2011] [Indexed: 11/24/2022]
Abstract
UBX domain is a general p97/VCP-binding module found in an increasing number of proteins including FAF1, p47, SAKS1 and UBXD7. FAF1, a multi-functional tumor suppressor protein, binds to the N domain of p97/VCP through its C-terminal UBX domain and thereby inhibits the proteasomal protein degradation in which p97/VCP acts as a co-chaperone. Here we report the crystal structure of human FAF1 UBX domain at 2.9Å resolution. It reveals that the conserved FP sequence in the p97/VCP-binding region adopts a rarely observed cis-Pro touch-turn structure. We call it an FcisP touch-turn motif and suggest that it is the conserved structural element of the UBX domain. Four FAF1 UBX molecules in an asymmetric unit of the crystal show two different conformations of the FcisP touch-turn motif. The phenyl ring of F(619) in the motif stacks partly over cis-Pro(620) in one conformation, whereas it is swung out from cis-P(620), in the other conformation, and forms hydrophobic contacts with the residues of the neighboring molecule. In addition, the entire FcisP touch-turn motif is pulled out in the second conformation by about 2Å in comparison to the first conformation. Those conformational differences observed in the p97/VCP-binding motif caused by the interaction with neighboring molecules presumably represent the conformational change of the UBX domain on its binding to the N domain of p97/VCP.
Collapse
|
38
|
Zhang L, Zhou F, van Laar T, Zhang J, van Dam H, Ten Dijke P. Fas-associated factor 1 antagonizes Wnt signaling by promoting β-catenin degradation. Mol Biol Cell 2011; 22:1617-24. [PMID: 21411632 PMCID: PMC3084683 DOI: 10.1091/mbc.e10-12-0985] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Fas-associated factor 1 (FAF1) is identified as a negative regulator of Wnt/β-catenin signaling. In addition, its involvement in osteoblast differentiation and mineralization is shown. FAF1 was found to promote β-catenin degradation by enhancing its polyubiquitination. The canonical Wnt pathway plays an important role in the regulation of cell proliferation and differentiation. Activation of this signaling pathway causes disruption of the Axin/adenomatous polyposis coli/glycogen synthase kinase 3β complex, resulting in stabilization of β-catenin and its association with lymphoid enhancer factor/T-cell factor in the nucleus. Here, we identify Fas-associated factor 1 (FAF1) as a negative regulator of Wnt/β-catenin signaling. We found overexpression of FAF1 to strongly inhibit Wnt-induced transcriptional reporter activity and to counteract Wnt-induced β-catenin accumulation. Moreover, knockdown of FAF1 resulted in an increase in β-catenin levels and in activation of Wnt/β-catenin–induced transcription. FAF1 was found to interact with β-catenin upon inhibition of proteasome. Ectopic expression of FAF1 promoted β-catenin degradation by enhancing its polyubiquitination. Functional studies in C2C12 myoblasts and KS483 preosteoblastic cells showed that FAF1 depletion resulted in activation of endogenous Wnt-induced genes and enhanced osteoblast differentiation, whereas FAF1 overexpression had the opposite effect. These results identify FAF1 as a novel inhibitory factor of canonical Wnt signaling pathway.
Collapse
Affiliation(s)
- Long Zhang
- Department of Molecular Cell Biology and Center for Biomedical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
39
|
Haines DS. p97-containing complexes in proliferation control and cancer: emerging culprits or guilt by association? Genes Cancer 2010; 1:753-763. [PMID: 21103003 DOI: 10.1177/1947601910381381] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
p97 (also called VCP in metazoans and CDC48 in yeast) is a highly conserved, abundant and essential type II ATPase that functions in numerous ubiquitin signaling dependent processes. p97/Cd48 activities require a growing number of adaptor or accessory proteins that promote interactions with ubiquitinated proteins. p97 has human disease relevance as it is mutated in familial cases of inclusion body myopathy associated with Paget's disease of the bone and frontotemporal dementia (IBMPFD). There is also increasing evidence suggesting that p97 and/or some of its adaptors play a role in cancer. This review will summarize our existing knowledge of the biochemical, molecular and cellular activities of p97-containing complexes, with an ending focus on their potential role in malignancy.
Collapse
Affiliation(s)
- Dale S Haines
- Fels Institute for Cancer Research and Molecular Biology and Department of Biochemistry, Temple University School of Medicine, Philadelphia, Pennsylvania, 19140, USA
| |
Collapse
|
40
|
Song J, Park JK, Lee JJ, Choi YS, Ryu KS, Kim JH, Kim E, Lee KJ, Jeon YH, Kim EE. Structure and interaction of ubiquitin-associated domain of human Fas-associated factor 1. Protein Sci 2010; 18:2265-76. [PMID: 19722279 DOI: 10.1002/pro.237] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Fas-associated factor (FAF)-1 is a multidomain protein that was first identified as a member of the Fas death-inducing signaling complex, but later found to be involved in various biological processes. Although the exact mechanisms are not clear, FAF1 seems to play an important role in cancer, asbestos-induced mesotheliomas, and Parkinson's disease. It interacts with polyubiquitinated proteins, Hsp70, and p97/VCP (valosin-containing protein), in addition to the proteins of the Fas-signaling pathway. We have determined the crystal structure of the ubiquitin-associated domain of human FAF1 (hFAF1-UBA) and examined its interaction with ubiquitin and ubiquitin-like proteins using nuclear magnetic resonance. hFAF1-UBA revealed a canonical three-helical bundle that selectively binds to mono- and di-ubiquitin (Lys48-linked), but not to SUMO-1 (small ubiquitin-related modifier 1) or NEDD8 (neural precursor cell expressed, developmentally down-regulated 8). The interaction between hFAF1-UBA and di-ubiquitin involves hydrophobic interaction accompanied by a transition in the di-ubiquitin conformation. These results provide structural insight into the mechanism of polyubiquitin recognition by hFAF1-UBA.
Collapse
Affiliation(s)
- Jinsue Song
- Magnetic Resonance Team, Korea Basic Science Institute, 804-1 Yangchung-Ri, Ochang, Chungbuk, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Kang W, Shin HY, Yang JK. Crystallization and preliminary X-ray crystallographic analysis of human FAF1 UBX domain. Acta Crystallogr Sect F Struct Biol Cryst Commun 2010; 66:211-3. [PMID: 20124726 PMCID: PMC2815696 DOI: 10.1107/s1744309110001077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Accepted: 01/08/2010] [Indexed: 11/10/2022]
Abstract
Fas-associated factor 1 (FAF1) is a multifunctional pro-apoptotic protein that is involved in Fas-mediated apoptosis, NF-kappaB signalling and the ubiquitin-proteasome pathway. In the ubiquitin-proteasome pathway, FAF1 binds to the N domain of p97/VCP, a molecular chaperone that acts in complex with the proteasome, through its C-terminal UBX domain and inhibits the proteasomal protein-degradation process. In an effort to elucidate the structural basis of the function of FAF1 in modulating p97/VCP activity related to proteasomal protein degradation, crystallographic analysis of the FAF1 UBX domain and the p97/VCP N domain was initiated. Following the recently reported crystallization of the FAF1 UBX domain bound to the p97/VCP N domain, the unbound FAF1 UBX domain was also crystallized for purposes of structural comparison. X-ray data were collected to 3.00 A resolution and the crystals belonged to space group F4(1)32, with unit-cell parameters a = b = c = 176.40 A. The Matthews coefficient and solvent content were estimated to be 3.04 A(3) Da(-1) and 59.5%, respectively, assuming that the asymmetric unit contained two molecules of the UBX domain, which was subsequently confirmed by molecular-replacement calculations.
Collapse
Affiliation(s)
- Wonchull Kang
- Department of Chemistry, Soongsil University, Seoul 156-743, Republic of Korea
| | - Hwa Young Shin
- Department of Chemistry, Soongsil University, Seoul 156-743, Republic of Korea
| | - Jin Kuk Yang
- Department of Chemistry, Soongsil University, Seoul 156-743, Republic of Korea
| |
Collapse
|
42
|
Shin HY, Kang W, Lee SY, Yang JK. Crystallization and preliminary X-ray crystallographic analysis of the N domain of p97/VCP in complex with the UBX domain of FAF1. Acta Crystallogr Sect F Struct Biol Cryst Commun 2010; 66:41-3. [PMID: 20057067 PMCID: PMC2805533 DOI: 10.1107/s1744309109047691] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2009] [Accepted: 11/11/2009] [Indexed: 05/28/2023]
Abstract
p97/VCP is a multifunctional AAA(+)-family ATPase that is involved in diverse cellular processes. p97/VCP directly interacts with various adaptors for activity in different biochemical contexts. Among these adaptors are p47 and Fas-associated factor 1 (FAF1), which contain a common UBX domain through which they bind to the N domain of p97/VCP. In the ubiquitin-proteasome pathway, p97/VCP acts as a chaperone that presents client proteins to the proteasome for degradation, while FAF1 modulates the process by interacting with ubiquitinated client proteins and also with p97/VCP. In an effort to elucidate the structural details of the interaction between p97/VCP and FAF1, the p97/VCP N domain was crystallized in complex with the FAF1 UBX domain. X-ray data were collected to 2.60 A resolution and the crystals belonged to space group C222(1), with unit-cell parameters a = 58.24, b = 72.81, c = 132.93 A. The Matthews coefficient and solvent content were estimated to be 2.39 A(3) Da(-1) and 48.4%, respectively, assuming that the asymmetric unit contained p97/VCP N domain and FAF1 molecules in a 1:1 ratio, which was subsequently confirmed by molecular-replacement calculations.
Collapse
Affiliation(s)
- Hwa Young Shin
- Department of Chemistry, College of Natural Sciences, Soongsil University, Seoul 156-743, Republic of Korea
| | - Wonchull Kang
- Department of Chemistry, College of Natural Sciences, Soongsil University, Seoul 156-743, Republic of Korea
| | - Sang Yoon Lee
- Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 443-721, Republic of Korea
| | - Jin Kuk Yang
- Department of Chemistry, College of Natural Sciences, Soongsil University, Seoul 156-743, Republic of Korea
| |
Collapse
|
43
|
Menges CW, Altomare DA, Testa JR. FAS-associated factor 1 (FAF1): diverse functions and implications for oncogenesis. Cell Cycle 2009; 8:2528-34. [PMID: 19597341 DOI: 10.4161/cc.8.16.9280] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
FAS-associated factor 1, FAF1, is an evolutionarily conserved protein that has several protein interaction domains. Although FAF1 was initially identified as a member of the FAS death-inducing signaling complex, subsequent work has revealed that FAF1 functions in diverse biological processes. FAF1 has been shown to play an important role in normal development and neuronal cell survival, whereas FAF1 downregulation may contribute to multiple aspects of tumorigenesis. In particular, there is compelling evidence implicating FAF1 as a tumor suppressor involved in the regulation of apoptosis and NFkappaB activity, as well as in ubiquitination and proteasomal degradation. Here, we highlight FAF1's role in NFkappaB signaling and postulate that this pathway has critical connotations for the pathogenesis and treatment of human cancers.
Collapse
Affiliation(s)
- Craig W Menges
- Human Genetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | |
Collapse
|
44
|
Activated TNF-alpha/NF-kappaB signaling via down-regulation of Fas-associated factor 1 in asbestos-induced mesotheliomas from Arf knockout mice. Proc Natl Acad Sci U S A 2009; 106:3420-5. [PMID: 19223589 DOI: 10.1073/pnas.0808816106] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The human CDKN2A locus encodes 2 distinct proteins, p16(INK4A) and p14(ARF) [mouse p19(Arf)], designated INK4A (inhibitor of cyclin dependent kinase 4) and ARF (alternative reading frame) here, that are translated from alternatively spliced mRNAs. Human ARF is implicated as a tumor suppressor gene, mainly in association with the simultaneous deletion of INK4A. However, questions remain as to whether loss of ARF alone is sufficient to drive tumorigenesis. Here, we report that mice deficient for Arf are susceptible to accelerated asbestos-induced malignant mesothelioma (MM). MMs arising in Arf (+/-) mice consistently exhibit biallelic inactivation of Arf, but, unexpectedly, do not acquire additional recurrent genetic alterations that we previously identified in asbestos-induced MMs arising in Nf2 (+/-) mice. Array CGH analysis was used to detect a recurrent deletion at chromosome 4C6 in MMs from Arf (+/-) mice. A candidate gene in this region, Faf1 (FAS-associated factor 1), was further explored, because it encodes a protein implicated in tumor cell survival and in the pathogenesis of some human tumor types. We confirmed hemizygous loss of Faf1 and down-regulation of Faf1 protein in a series of MMs from Arf (+/-) mice, and we then showed that Faf1 regulates TNF-alpha-mediated NF-kappaB signaling, a pathway previously implicated in asbestos-induced oncogenesis of human mesothelial cells. Collectively, these data indicate that Arf inactivation has a significant role in driving MM pathogenesis, and implicate Faf1 as a key component in the TNF-alpha/NF-kappaB signaling node that has now been independently implicated in asbestos-induced oncogenesis.
Collapse
|
45
|
Uniparental disomies, homozygous deletions, amplifications, and target genes in mantle cell lymphoma revealed by integrative high-resolution whole-genome profiling. Blood 2008; 113:3059-69. [PMID: 18984860 DOI: 10.1182/blood-2008-07-170183] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mantle cell lymphoma (MCL) is genetically characterized by the t(11;14)(q13;q32) translocation and a high number of secondary chromosomal alterations. However, only a limited number of target genes have been identified. We have studied 10 MCL cell lines and 28 primary tumors with a combination of a high-density single-nucleotide polymorphism array and gene expression profiling. We detected highly altered genomes in the majority of the samples with a high number of partial uniparental disomies (UPDs). The UPD at 17p was one of the most common, and it was associated with TP53 gene inactivation. Homozygous deletions targeted 4 known tumor suppressor genes (CDKN2C, BCL2L11, CDKN2A, and RB1) and 6 new genes (FAF1, MAP2, SP100, MOBKL2B, ZNF280A, and PRAME). Gene amplification coupled with overexpression was identified in 35 different regions. The most recurrent amplified regions were 11q13.3-q13.5, 13q31.3, and 18q21.33, which targeted CCND1, C13orf25, and BCL2, respectively. Interestingly, the breakpoints flanking all the genomic alterations, including UPDs, were significantly associated with genomic regions enriched in copy number variants and segmental duplications, suggesting that the recombination at these regions may play a role in the genomic instability of MCL. This integrative genomic analysis has revealed target genes that may be potentially relevant in MCL pathogenesis.
Collapse
|
46
|
Jang MS, Sul JW, Choi BJ, Lee SJ, Suh JH, Kim NS, Kim WH, Lim DS, Lee CW, Kim E. Negative Feedback Regulation of Aurora-A via Phosphorylation of Fas-associated Factor-1. J Biol Chem 2008; 283:32344-51. [DOI: 10.1074/jbc.m804199200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|