1
|
Li YE, Liu S, Wang L, Du Y, Wu L, Chen H, Zhu T, Lin J, Xiong S, Wang Y, Zheng Q, Zou R, Lin L, Li Z, Wang L, Ge J, Ren J, Zhang Y. March2 Alleviates Aortic Aneurysm/Dissection by Regulating PKM2 Polymerization. Circ Res 2025; 136:e73-e93. [PMID: 40079144 DOI: 10.1161/circresaha.124.325049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 02/19/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Aortic aneurysm/dissection (AAD) is a life-threatening disease lacking effective pharmacological treatment. Protein ubiquitination plays a pivotal role in cardiovascular diseases. However, the possible contribution of the E3 ubiquitin ligase March2 (membrane-associated RING [really interesting new gene] finger protein 2) to the cause of AAD remains elusive. METHODS Integrated single-cell RNA sequencing analysis was conducted in human AAD tissues. Based on the screening results, we generated a mouse line of smooth muscle cell-specific March2 knockout. β-Aminopropionitrile monofumarate was used to establish AAD. Cleavage under targets and tagmentation and cleavage under targets and tagmentation-quantitative polymerase chain reaction were performed to identify possible target genes for histone H3K18 lactylation. RESULTS March2 expression was downregulated in aorta from patients with AAD or β-aminopropionitrile monofumarate-induced AAD mice. β-Aminopropionitrile monofumarate-induced AAD was significantly accentuated in March2 global (March2-/-) and vascular smooth muscle cell-specific deletion (March2fl/fl; TaglnCre) mice, whereas the AAD pathology was rescued by rAAV9-SM22α (smooth muscle 22α)-March2 (recombinant adeno-associated virus serotype 9 expressing Flag-tagged March2 under SM22α promoter). March2 interacted with PKM2 (pyruvate kinase M2) to promote K33-linked polyubiquitination. Deficiency of March2 lessened PKM2 dimer-to-tetramer conversion in AAD and overtly exacerbated AAD-induced histone H3K18 lactylation in vascular smooth muscle cells by fostering glucose metabolism reprogramming, thereby promoting p53-driven apoptotic transcriptional response-a hallmark of AAD pathogenesis. TEPP-46 (tetraethyl pyrophosphate), a PKM2-specific activator, pronouncedly alleviated March2 deficiency-deteriorated AAD pathology. CONCLUSIONS Our findings demonstrated that March2 is a novel endogenous defender that prevents AAD by inhibiting vascular smooth muscle cell apoptosis, suggesting that March2 represents a potential therapeutic target for AAD.
Collapse
Affiliation(s)
- Yiran E Li
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Shuolin Liu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- Department of Vascular Surgery (Z.L., Lixin Wang), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Litao Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Yuxin Du
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Lin Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Haoran Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Tingfang Zhu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Jie Lin
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Shengjun Xiong
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Yayu Wang
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Guangdong, China (Y.W., Q.Z.)
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, Guangdong (Y.W.)
| | - Qijun Zheng
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Guangdong, China (Y.W., Q.Z.)
| | - Rongjun Zou
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine (R.Z.)
| | - Ling Lin
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Zheyun Li
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- Department of Vascular Surgery (Z.L., Lixin Wang), Zhongshan Hospital, Fudan University, China
- Vascular Surgery Institute of Fudan University, Shanghai, China (Z.L., Lixin Wang)
| | - Lixin Wang
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- Department of Vascular Surgery (Z.L., Lixin Wang), Zhongshan Hospital, Fudan University, China
- Vascular Surgery Institute of Fudan University, Shanghai, China (Z.L., Lixin Wang)
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| | - Yingmei Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- State Key Laboratory of Cardiovascular Diseases (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., Z.L., Lixin Wang, J.G., J.R., Y.Z.), Zhongshan Hospital, Fudan University, China
- National Health Commission (NHC) Key Laboratory of Ischemic Heart Diseases, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- National Clinical Research Center for Interventional Medicine, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (Y.E.L., S.L., Litao Wang, Y.D., L. Wu, H.C., T.Z., J.L., S.X., L.L., J.G., J.R., Y.Z.)
| |
Collapse
|
2
|
Jayamali BPMV, Wijerathna HMSM, Sirisena DMKP, Hanchapola HACR, Warnakula WADLR, Arachchi UPE, Liyanage DS, Jung S, Wan Q, Lee J. Molecular depiction and functional delineation of E3 ubiquitin ligase MARCH5 in yellowtail clownfish (Amphiprion clarkii). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 162:105283. [PMID: 39481581 DOI: 10.1016/j.dci.2024.105283] [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: 05/23/2024] [Revised: 10/10/2024] [Accepted: 10/27/2024] [Indexed: 11/02/2024]
Abstract
Membrane-associated Ring-CH 5 (MARCH5) is a mitochondrial E3 ubiquitin ligase playing a key role in the regulation of mitochondrial dynamics. In mammals, MARCH5 negatively regulates mitochondrial antiviral signaling (MAVS) protein aggregation during viral infection and hampers downstream type I interferon signaling to prevent excessive immune activation. However, its precise functional role in the teleost immune system remains unclear. This study investigated the molecular characteristics and immune response of the MARCH5 ortholog in Amphiprion clarkii (A. clarkii; AcMARCH5). The predicted AcMARCH5 protein sequence consists of 287 amino acids with a molecular weight of 32.02 kDa and a theoretical isoelectric point of 9.11. It contains four C-terminal transmembrane (TM) domains and an N-terminal RING cysteine-histidine (CH) domain, which directly regulates ubiquitin transfer. Multiple sequence alignment revealed a high level of conservation between AcMARCH5 and its orthologs in other vertebrate species. Under normal physiological conditions, AcMARCH5 showed the highest mRNA expression in the muscle, brain, and kidney tissues of A. clarkii. Upon stimulation with polyinosinic:polycytidylic acid (Poly I:C), lipopolysaccharide (LPS), and Vibrio harveyi, AcMARCH5 expression was drastically modulated. Functional assays showed that overexpression of AcMARCH5 in fathead minnow (FHM) cells downregulated antiviral gene expression, accompanied by enhanced viral hemorrhagic septicemia virus (VHSV) replication. In murine macrophages, AcMARCH5 overexpression markedly reduced the production of pro-inflammatory cytokines in response to poly I:C treatment. Additionally, AcMARCH5 exhibited an anti-apoptotic effect in H2O2-treated FHM cells. Collectively, these results suggest that AcMARCH5 may play a role in maintaining cellular homeostasis under disease and stress conditions in A. clarkii.
Collapse
Affiliation(s)
- B P M Vileka Jayamali
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - H M S M Wijerathna
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - D M K P Sirisena
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - H A C R Hanchapola
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - W A D L R Warnakula
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - U P E Arachchi
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - D S Liyanage
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Sumi Jung
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Kidang Marine Science Institute of Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - Qiang Wan
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Kidang Marine Science Institute of Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea.
| | - Jehee Lee
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Kidang Marine Science Institute of Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea.
| |
Collapse
|
3
|
Behera A, Sachan D, Barik GK, Reddy ABM. Role of MARCH E3 ubiquitin ligases in cancer development. Cancer Metastasis Rev 2024; 43:1257-1277. [PMID: 39037545 DOI: 10.1007/s10555-024-10201-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
Membrane-associated RING-CH (MARCH) E3 ubiquitin ligases, a family of RING-type E3 ubiquitin ligases, have garnered increased attention for their indispensable roles in immune regulation, inflammation, mitochondrial dynamics, and lipid metabolism. The MARCH E3 ligase family consists of eleven distinct members, and the dysregulation of many of these members has been documented in several human malignancies. Over the past two decades, extensive research has revealed that MARCH E3 ligases play pivotal roles in cancer progression by ubiquitinating key oncogenes and tumor suppressors and orchestrating various signaling pathways. Some MARCH E3s act as oncogenes, while others act as tumor suppressors, and the majority of MARCH E3s play both oncogenic and tumor suppressive roles in a context-dependent manner. Notably, there is special emphasis on the sole mitochondrial MARCH E3 ligase MARCH5, which regulates mitochondrial homeostasis within cancer cells. In this review, we delve into the diverse functions of MARCH E3 ligases across different cancer types, shedding light on the underlying molecular mechanisms mediating their effects, their regulatory effects on cancer and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Abhayananda Behera
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Deepanshi Sachan
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Ganesh Kumar Barik
- Cancer Biology Division, National Centre for Cell Science, Savitribai Phule Pune University, Ganeshkhind Road, Pune, Maharashtra, 411007, India
| | | |
Collapse
|
4
|
Umthong S, Timilsina U, D’Angelo MR, Salka K, Stavrou S. MARCH2, a T cell specific factor that restricts HIV-1 infection. PLoS Pathog 2024; 20:e1012330. [PMID: 39074162 PMCID: PMC11309421 DOI: 10.1371/journal.ppat.1012330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 08/08/2024] [Accepted: 06/07/2024] [Indexed: 07/31/2024] Open
Abstract
Membrane-associated RING-CH (MARCH) 2 is a member of the MARCH protein family of RING-CH finger E3 ubiquitin ligases that play important roles in regulating the levels of proteins found on the cell surface. MARCH1, 2 and 8 inhibit HIV-1 infection by preventing the incorporation of the envelope glycoproteins into nascent virions. However, a better understanding of the mechanism utilized by MARCH proteins to restrict HIV-1 infection is needed. In this report, we identify an amino acid in human MARCH2, absent in mouse MARCH2, critical for its antiretroviral function. Moreover, we map the domains of human MARCH2 critical for restricting as well as binding to the HIV-1 envelope glycoproteins. In addition, we demonstrate that MARCH2 is present inside nascent virions and reduces particle infectivity by blocking virus entry in a RING-CH-independent manner. Finally, we show that MARCH2 acts as an HIV-1 restriction factor only in primary CD4+ T cells and can prevent cell-to-cell transmission of HIV-1. Our findings reveal important new aspects of the antiviral mechanism utilized by human MARCH2 to restrict HIV-1 that have potential implications to all MARCH proteins with antiviral functions and their viral targets.
Collapse
Affiliation(s)
- Supawadee Umthong
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Uddhav Timilsina
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
| | - Mary R. D’Angelo
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
| | - Kyle Salka
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
| | - Spyridon Stavrou
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
| |
Collapse
|
5
|
Saferali A, Kim W, Xu Z, Chase RP, Cho MH, Laederach A, Castaldi PJ, Hersh CP. Colocalization analysis of 3' UTR alternative polyadenylation quantitative trait loci reveals novel mechanisms underlying associations with lung function. Hum Mol Genet 2024; 33:1164-1175. [PMID: 38569558 DOI: 10.1093/hmg/ddae055] [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: 09/18/2023] [Revised: 02/02/2024] [Indexed: 04/05/2024] Open
Abstract
While many disease-associated single nucleotide polymorphisms (SNPs) are expression quantitative trait loci (eQTLs), a large proportion of genome-wide association study (GWAS) variants are of unknown function. Alternative polyadenylation (APA) plays an important role in posttranscriptional regulation by allowing genes to shorten or extend 3' untranslated regions (UTRs). We hypothesized that genetic variants that affect APA in lung tissue may lend insight into the function of respiratory associated GWAS loci. We generated alternative polyadenylation (apa) QTLs using RNA sequencing and whole genome sequencing on 1241 subjects from the Lung Tissue Research Consortium (LTRC) as part of the NHLBI TOPMed project. We identified 56 179 APA sites corresponding to 13 582 unique genes after filtering out APA sites with low usage. We found that a total of 8831 APA sites were associated with at least one SNP with q-value < 0.05. The genomic distribution of lead APA SNPs indicated that the majority are intronic variants (33%), followed by downstream gene variants (26%), 3' UTR variants (17%), and upstream gene variants (within 1 kb region upstream of transcriptional start site, 10%). APA sites in 193 genes colocalized with GWAS data for at least one phenotype. Genes containing the top APA sites associated with GWAS variants include membrane associated ring-CH-type finger 2 (MARCHF2), nectin cell adhesion molecule 2 (NECTIN2), and butyrophilin subfamily 3 member A2 (BTN3A2). Overall, these findings suggest that APA may be an important mechanism for genetic variants in lung function and chronic obstructive pulmonary disease (COPD).
Collapse
Affiliation(s)
- Aabida Saferali
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, United States
| | - Wonji Kim
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, United States
| | - Zhonghui Xu
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, United States
| | - Robert P Chase
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, United States
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, United States
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115, United States
| | - Alain Laederach
- Department of Biology, University of North Carolina at Chapel Hill, 120 South Road, Chapel Hill, NC 27599, United States
| | - Peter J Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, United States
- Division of General Medicine and Primary Care, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, United States
| | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, United States
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115, United States
| |
Collapse
|
6
|
Ito K, Harada I, Martinez C, Sato K, Lee E, Port E, Byerly JH, Nayak A, Tripathi E, Zhu J, Irie HY. MARCH2, a Novel Oncogene-regulated SNAIL E3 Ligase, Suppresses Triple-negative Breast Cancer Metastases. CANCER RESEARCH COMMUNICATIONS 2024; 4:946-957. [PMID: 38457262 PMCID: PMC10977041 DOI: 10.1158/2767-9764.crc-23-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 01/02/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024]
Abstract
Epithelial-mesenchymal transition (EMT) in cancer promotes metastasis and chemotherapy resistance. A subset of triple-negative breast cancer (TNBC) exhibits a mesenchymal gene signature that is associated with poor patient outcomes. We previously identified PTK6 tyrosine kinase as an oncogenic driver of EMT in a subset of TNBC. PTK6 induces EMT by stabilizing SNAIL, a key EMT-initiating transcriptional factor. Inhibition of PTK6 activity reverses mesenchymal features of TNBC cells and suppresses their metastases by promoting SNAIL degradation via a novel mechanism. In the current study, we identify membrane-associated RING-CH2 (MARCH2) as a novel PTK6-regulated E3 ligase that promotes the ubiquitination and degradation of SNAIL protein. The MARCH2 RING domain is critical for SNAIL ubiquitination and subsequent degradation. PTK6 inhibition promotes the interaction of MARCH2 with SNAIL. Overexpression of MARCH2 exhibits tumor suppressive properties and phenocopies the effects of SNAIL downregulation and PTK6 inhibition in TNBC cells, such as inhibition of migration, anoikis resistance, and metastasis. Consistent with this, higher levels of MARCH2 expression in breast and other cancers are associated with better prognosis. We have identified MARCH2 as a novel SNAIL E3 ligase that regulates EMT and metastases of mesenchymal TNBC. SIGNIFICANCE EMT is a process directly linked to drug resistance and metastasis of cancer cells. We identified MARCH2 as a novel regulator of SNAIL, a key EMT driver, that promotes SNAIL ubiquitination and degradation in TNBC cells. MARCH2 is oncogene regulated and inhibits growth and metastasis of TNBC. These insights could contribute to novel strategies to therapeutically target TNBC.
Collapse
Affiliation(s)
- Koichi Ito
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ibuki Harada
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Criseyda Martinez
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Katsutoshi Sato
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Elisa Port
- Department of Surgery, Mount Sinai Hospital, New York, New York
| | - Jessica H. Byerly
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Anupma Nayak
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ekta Tripathi
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jun Zhu
- Sema4, Stamford, Connecticut
| | - Hanna Y. Irie
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
7
|
Liu S, Bi Y, Han T, Li YE, Wang Q, Wu NN, Xu C, Ge J, Hu R, Zhang Y. The E3 ubiquitin ligase MARCH2 protects against myocardial ischemia-reperfusion injury through inhibiting pyroptosis via negative regulation of PGAM5/MAVS/NLRP3 axis. Cell Discov 2024; 10:24. [PMID: 38409220 PMCID: PMC10897310 DOI: 10.1038/s41421-023-00622-3] [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: 03/29/2023] [Accepted: 09/12/2023] [Indexed: 02/28/2024] Open
Abstract
Inflammasome activation and pyroptotic cell death are known to contribute to the pathogenesis of cardiovascular diseases, such as myocardial ischemia-reperfusion (I/R) injury, although the underlying regulatory mechanisms remain poorly understood. Here we report that expression levels of the E3 ubiquitin ligase membrane-associated RING finger protein 2 (MARCH2) were elevated in ischemic human hearts or mouse hearts upon I/R injury. Genetic ablation of MARCH2 aggravated myocardial infarction and cardiac dysfunction upon myocardial I/R injury. Single-cell RNA-seq analysis suggested that loss of MARCH2 prompted activation of NLRP3 inflammasome in cardiomyocytes. Mechanistically, phosphoglycerate mutase 5 (PGAM5) was found to act as a novel regulator of MAVS-NLRP3 signaling by forming liquid-liquid phase separation condensates with MAVS and fostering the recruitment of NLRP3. MARCH2 directly interacts with PGAM5 to promote its K48-linked polyubiquitination and proteasomal degradation, resulting in reduced PGAM5-MAVS co-condensation, and consequently inhibition of NLRP3 inflammasome activation and cardiomyocyte pyroptosis. AAV-based re-introduction of MARCH2 significantly ameliorated I/R-induced mouse heart dysfunction. Altogether, our findings reveal a novel mechanism where MARCH2-mediated ubiquitination negatively regulates the PGAM5/MAVS/NLRP3 axis to protect against cardiomyocyte pyroptosis and myocardial I/R injury.
Collapse
Affiliation(s)
- Shuolin Liu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Key Laboratory of Viral Heart Diseases, National Health Commission. Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yaguang Bi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Key Laboratory of Viral Heart Diseases, National Health Commission. Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Tianting Han
- College of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai, China
- State Key Laboratory of Molecular Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Yiran E Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Key Laboratory of Viral Heart Diseases, National Health Commission. Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Qihang Wang
- College of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai, China
- State Key Laboratory of Molecular Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Ne Natalie Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Key Laboratory of Viral Heart Diseases, National Health Commission. Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Chenguo Xu
- College of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Key Laboratory of Viral Heart Diseases, National Health Commission. Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
| | - Ronggui Hu
- College of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai, China.
- State Key Laboratory of Molecular Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Life Science, Hangzhou Institute for Advance Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Key Laboratory of Viral Heart Diseases, National Health Commission. Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
| |
Collapse
|
8
|
Yu C, Bai Y, Tan W, Bai Y, Li X, Zhou Y, Zhai J, Xue M, Tang YD, Zheng C, Liu Q. Human MARCH1, 2, and 8 block Ebola virus envelope glycoprotein cleavage via targeting furin P domain. J Med Virol 2024; 96:e29445. [PMID: 38299743 DOI: 10.1002/jmv.29445] [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: 05/09/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/02/2024]
Abstract
Membrane-associated RING-CH (MARCH) family proteins were recently reported to inhibit viral replication through multiple modes. Previous work showed that human MARCH8 blocked Ebola virus (EBOV) glycoprotein (GP) maturation. Our study here demonstrates that human MARCH1 and MARCH2 share a similar pattern to MARCH8 in restricting EBOV GP-pseudotyped viral infection. Human MARCH1 and MARCH2 retain EBOV GP at the trans-Golgi network, reduce its cell surface display, and impair EBOV GP-pseudotyped virions infectivity. Furthermore, we uncover that the host proprotein convertase furin could interact with human MARCH1/2 and EBOV GP intracellularly. Importantly, the furin P domain is verified to be recognized by MARCH1/2/8, which is critical for their blocking activities. Besides, bovine MARCH2 and murine MARCH1 also impair EBOV GP proteolytic processing. Altogether, our findings confirm that MARCH1/2 proteins of different mammalian origins showed a relatively conserved feature in blocking EBOV GP cleavage, which could provide clues for subsequent MARCHs antiviral studies and may facilitate the development of novel strategies to antagonize enveloped virus infection.
Collapse
Affiliation(s)
- Changqing Yu
- Engineering Center of Agricultural Biosafety Assessment and Biotechnology, School of Advanced Agricultural Sciences, Yibin Vocational and Technical College, Yibin, People's Republic of China
| | - Yuanzhe Bai
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Wenbo Tan
- College of Advanced Agriculture and Ecological Environment, Heilongjiang University, Harbin, People's Republic of China
| | - Yu Bai
- College of Animal Science, Wenzhou Vocational College of Science and Technology, Wenzhou, People's Republic of China
| | - Xuemei Li
- Engineering Center of Agricultural Biosafety Assessment and Biotechnology, School of Advanced Agricultural Sciences, Yibin Vocational and Technical College, Yibin, People's Republic of China
| | - Yulong Zhou
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, People's Republic of China
| | - Jingbo Zhai
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao, People's Republic of China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yan-Dong Tang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Chunfu Zheng
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, People's Republic of China
- Department of Microbiology, Immunology & Infection Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Qiang Liu
- Nanchong Key Laboratory of Disease Prevention, Control and Detection in Livestock and Poultry, Nanchong Vocational and Technical College, Nanchong, People's Republic of China
| |
Collapse
|
9
|
Yu C, Wang G, Liu Q, Zhai J, Xue M, Li Q, Xian Y, Zheng C. Host antiviral factors hijack furin to block SARS-CoV-2, ebola virus, and HIV-1 glycoproteins cleavage. Emerg Microbes Infect 2023; 12:2164742. [PMID: 36591809 PMCID: PMC9897805 DOI: 10.1080/22221751.2022.2164742] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Viral envelope glycoproteins are crucial for viral infections. In the process of enveloped viruses budding and release from the producer cells, viral envelope glycoproteins are presented on the viral membrane surface as spikes, promoting the virus's next-round infection of target cells. However, the host cells evolve counteracting mechanisms in the long-term virus-host co-evolutionary processes. For instance, the host cell antiviral factors could potently suppress viral replication by targeting their envelope glycoproteins through multiple channels, including their intracellular synthesis, glycosylation modification, assembly into virions, and binding to target cell receptors. Recently, a group of studies discovered that some host antiviral proteins specifically recognized host proprotein convertase (PC) furin and blocked its cleavage of viral envelope glycoproteins, thus impairing viral infectivity. Here, in this review, we briefly summarize several such host antiviral factors and analyze their roles in reducing furin cleavage of viral envelope glycoproteins, aiming at providing insights for future antiviral studies.
Collapse
Affiliation(s)
- Changqing Yu
- School of Advanced Agricultural Sciences, Yibin Vocational and Technical College, Yibin, People’s Republic of China
| | - Guosheng Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Qiang Liu
- Nanchong Key Laboratory of Disease Prevention, Control and Detection in Livestock and Poultry, Nanchong Vocational and Technical College, Nanchong, People’s Republic of China
| | - Jingbo Zhai
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao, People’s Republic of China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China,Mengzhou Xue
| | - Qiang Li
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China,Qiang Li
| | - Yuanhua Xian
- School of Advanced Agricultural Sciences, Yibin Vocational and Technical College, Yibin, People’s Republic of China,Yuanhua Xian
| | - Chunfu Zheng
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, People’s Republic of China,Department of Microbiology, Immunology & Infection Diseases, University of Calgary, Calgary, Canada, Chunfu Zheng
| |
Collapse
|
10
|
Umthong S, Timilsina U, D’Angelo M, Stavrou S. Determining the antiviral mechanism of MARCH2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.18.558306. [PMID: 37786722 PMCID: PMC10541590 DOI: 10.1101/2023.09.18.558306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Membrane-associated RING-CH (MARCH) 2 protein is a member of the MARCH protein family of RING-CH finger E3 ubiquitin ligases that have important functions in regulating the levels of proteins found on the cell surface. MARCH1, 2 and 8 inhibit HIV-1 infection by preventing the incorporation of the envelope glycoproteins in nascent virions. However, a better understanding on the mechanism utilized by MARCH proteins to restrict HIV-1 is needed. In this report, we identify an amino acid in human MARCH2, that is absent in mouse MARCH2, critical for its antiretroviral function. Moreover, we map the domains of human MARCH2 critical for restricting as well as binding to the HIV-1 envelope glycoproteins. Our findings reveal important new aspects of the antiviral mechanism utilized by human MARCH2 to restrict HIV-1 that have potential implications to all MARCH proteins with antiviral functions.
Collapse
Affiliation(s)
- Supawadee Umthong
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Uddhav Timilsina
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Mary D’Angelo
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Spyridon Stavrou
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
11
|
Tang X, Wang Z, Jiang D, Chen M, Zhang D. Expression profile, subcellular localization of MARCH4 and transcriptome analysis of its potential regulatory signaling pathway in large yellow croaker (Larimichthys crocea). FISH & SHELLFISH IMMUNOLOGY 2022; 130:273-282. [PMID: 36126839 DOI: 10.1016/j.fsi.2022.09.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/22/2022] [Accepted: 09/10/2022] [Indexed: 06/15/2023]
Abstract
Membrane-associated RING-CH (MARCH) family, as Ring-type E3 ligases, have attracted extensive attention to their immune functions. MARCH4 plays an essential role in regulating immune response in mammal. In the present study, it is the first to report on MARCH4 characteristics and signal pathway in fish. MARCH4 in large yellow croaker Larimichthys crocea (named as LcMARCH4) encodes a RING-CH domain and two TM domains, as well as other function domains, including an N-terminal proline rich domain, an AxxxG-motif in TM1, a tyrosine-based YXXØ motif, and a C-terminal PDZ-binding domain. LcMARCH4 is a tissue-specific protein with highly significant expression in brain. The mRNA transcripts of LcMARCH4 were significantly induced in the main organs (skin, gill, spleen, and head-kidney) by C. irritans infection. Consistently, significant increase was observed in spleen and head-kidney after LPS, Poly I:C stimulation and V. parahaemolyticus infection. Subcellular localization analysis showed that LcMARCH4 was localized in the cytoplasm and membrane. Moreover, we found 46 DEGs in a comparative transcriptome analysis between the LcMARCH4 overexpression group and control vector group. The analysis showed that HSPA6, HSPA1B and DNAJB1 might play important regulatory roles to MARCH4 in fish. Notably, two noncoding RNA, both RN7SL1 and RN7SL2, the expression levels went up in MARCH4 overexpression cells. Taken together, this study will provide new insights into finfish MARCH4 and its potential regulatory signaling pathway as well.
Collapse
Affiliation(s)
- Xin Tang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China
| | - Zhiyong Wang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Dan Jiang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China
| | - Meiling Chen
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China
| | - Dongling Zhang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China.
| |
Collapse
|
12
|
Alexander GM, Heiman-Patterson TD, Bearoff F, Sher RB, Hennessy L, Terek S, Caccavo N, Cox GA, Philip VM, Blankenhorn EA. Identification of quantitative trait loci for survival in the mutant dynactin p150Glued mouse model of motor neuron disease. PLoS One 2022; 17:e0274615. [PMID: 36107978 PMCID: PMC9477371 DOI: 10.1371/journal.pone.0274615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/01/2022] [Indexed: 11/19/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common degenerative motor neuron disorder. Although most cases of ALS are sporadic, 5-10% of cases are familial, with mutations associated with over 40 genes. There is variation of ALS symptoms within families carrying the same mutation; the disease may develop in one sibling and not in another despite the presence of the mutation in both. Although the cause of this phenotypic variation is unknown, it is likely related to genetic modifiers of disease expression. The identification of ALS causing genes has led to the development of transgenic mouse models of motor neuron disease. Similar to families with familial ALS, there are background-dependent differences in disease phenotype in transgenic mouse models of ALS suggesting that, as in human ALS, differences in phenotype may be ascribed to genetic modifiers. These genetic modifiers may not cause ALS rather their expression either exacerbates or ameliorates the effect of the mutant ALS causing genes. We have reported that in both the G93A-hSOD1 and G59S-hDCTN1 mouse models, SJL mice demonstrated a more severe phenotype than C57BL6 mice. From reciprocal intercrosses between G93A-hSOD1 transgenic mice on SJL and C57BL6 strains, we identified a major quantitative trait locus (QTL) on mouse chromosome 17 that results in a significant shift in lifespan. In this study we generated reciprocal intercrosses between transgenic G59S-hDCTN1 mice on SJL and C57BL6 strains and identified survival QTLs on mouse chromosomes 17 and 18. The chromosome 17 survival QTL on G93A-hSOD1 and G59S-hDCTN1 mice partly overlap, suggesting that the genetic modifiers located in this region may be shared by these two ALS models despite the fact that motor neuron degeneration is caused by mutations in different proteins. The overlapping region contains eighty-seven genes with non-synonymous variations predicted to be deleterious and/or damaging. Two genes in this segment, NOTCH3 and Safb/SAFB1, have been associated with motor neuron disease. The identification of genetic modifiers of motor neuron disease, especially those modifiers that are shared by SOD1 and dynactin-1 transgenic mice, may result in the identification of novel targets for therapies that can alter the course of this devastating illness.
Collapse
Affiliation(s)
| | - Terry D. Heiman-Patterson
- Department of Neurology, Lewis Katz School of Medicine of Temple University, Philadelphia, Pennsylvania, United States of America
| | - Frank Bearoff
- Department of Microbiology Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Roger B. Sher
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York, United States of America
| | - Laura Hennessy
- The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Shannon Terek
- The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Nicole Caccavo
- Department of Neurology, Lewis Katz School of Medicine of Temple University, Philadelphia, Pennsylvania, United States of America
| | - Gregory A. Cox
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Vivek M. Philip
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Elizabeth A. Blankenhorn
- Department of Microbiology Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
13
|
Smuggle tau through a secret(ory) pathway. Biochem J 2021; 478:2921-2925. [PMID: 34319403 DOI: 10.1042/bcj20210324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 11/17/2022]
Abstract
Secretion of misfolded tau, a microtubule-binding protein enriched in nerve cells, is linked to the progression of tau pathology. However, the molecular mechanisms underlying tau secretion are poorly understood. Recent work by Lee et al. [Biochemical J. (2021) 478: 1471-1484] demonstrated that the transmembrane domains of syntaxin6 and syntaxin8 could be exploited for tau release, setting a stage for testing a novel hypothesis that has profound implications in tauopathies (e.g. Alzheimer's disease, FTDP-17, and CBD/PSP) and other related neurodegenerative diseases. The present commentary highlights the importance and limitations of the study, and discusses opportunities and directions for future investigations.
Collapse
|
14
|
Abstract
Viral envelope glycoproteins are an important structural component on the surfaces of enveloped viruses that direct virus binding and entry and also serve as targets for the host adaptive immune response. In this study, we investigate the mechanism of action of the MARCH family of cellular proteins that disrupt the trafficking and virion incorporation of viral glycoproteins across several virus families. An emerging class of cellular inhibitory proteins has been identified that targets viral glycoproteins. These include the membrane-associated RING-CH (MARCH) family of E3 ubiquitin ligases that, among other functions, downregulate cell surface proteins involved in adaptive immunity. The RING-CH domain of MARCH proteins is thought to function by catalyzing the ubiquitination of the cytoplasmic tails (CTs) of target proteins, leading to their degradation. MARCH proteins have recently been reported to target retroviral envelope glycoproteins (Env) and vesicular stomatitis virus G glycoprotein (VSV-G). However, the mechanism of antiviral activity remains poorly defined. Here we show that MARCH8 antagonizes the full-length forms of HIV-1 Env, VSV-G, Ebola virus glycoprotein (EboV-GP), and the spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), thereby impairing the infectivity of virions pseudotyped with these viral glycoproteins. This MARCH8-mediated targeting of viral glycoproteins requires the E3 ubiquitin ligase activity of the RING-CH domain. We observe that MARCH8 protein antagonism of VSV-G is CT dependent. In contrast, MARCH8-mediated targeting of HIV-1 Env, EboV-GP, and SARS-CoV-2 S protein by MARCH8 does not require the CT, suggesting a novel mechanism of MARCH-mediated antagonism of these viral glycoproteins. Confocal microscopy data demonstrate that MARCH8 traps the viral glycoproteins in an intracellular compartment. We observe that the endogenous expression of MARCH8 in several relevant human cell types is rapidly inducible by type I interferon. These results help to inform the mechanism by which MARCH proteins exert their antiviral activity and provide insights into the role of cellular inhibitory factors in antagonizing the biogenesis, trafficking, and virion incorporation of viral glycoproteins.
Collapse
|
15
|
Ishihara T, Kanon H, Ban-Ishihara R, Ishihara N. Multiple assay systems to analyze the dynamics of mitochondrial nucleoids in living mammalian cells. Biochim Biophys Acta Gen Subj 2021; 1865:129874. [PMID: 33607223 DOI: 10.1016/j.bbagen.2021.129874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 01/18/2021] [Accepted: 02/09/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND Mitochondria, which play a critical role in energy production by oxidative respiration, are highly dynamic organelles and their double membranes undergo frequent events of fusion and fission. Mitochondria are believed to be derived from the endosymbiosis of proteobacteria, and thus mitochondria still contain their own DNA, mitochondrial DNA (mtDNA). Several copies of mtDNA form mitochondrial nucleoid with DNA-binding proteins. Recently, the morphology and distribution of the mitochondrial membrane and nucleoid were reported to be cooperatively regulated during their dynamic movement. However, the molecular mechanism is unclear, because the involved molecules are poorly understood, and suitable techniques to analyze nucleoid have not been fully developed. RESULTS To solve these issues, we examined the molecular mechanism of nucleoid dynamics by two approaches. First, we constructed a new probe to perform live imaging of nucleoid dynamics using the DNA-binding domain of mitochondrial transcriptional factor A (TFAM) and the photo-convertible fluorescent protein Kikume Green-Red (KikGR). Nucleoids were visualized stably for a long period using the new probe. Second, we searched for nucleoid regulatory factors by small interfering RNA screening using HeLa cells and identified a subset of MARCH family ubiquitin ligases that affect nucleoid morphology. CONCLUSION The factors and probe, reported in this study, would be useful to reveal novel mechanisms of mitochondrial regulation. GENERAL SIGNIFICANCE The mtDNA dynamics should be concerned in the regulation of mitochondrial activity and its quality control, associated with mitochondrial membrane dynamics.
Collapse
Affiliation(s)
- Takaya Ishihara
- Department of Biological Sciences, Graduate School of Science, Osaka University, Japan; Department of Protein Biochemistry, Institute of Life Science, Kurume University, Japan.
| | - Hirotaka Kanon
- Department of Biological Sciences, Graduate School of Science, Osaka University, Japan
| | - Reiko Ban-Ishihara
- Department of Protein Biochemistry, Institute of Life Science, Kurume University, Japan
| | - Naotada Ishihara
- Department of Biological Sciences, Graduate School of Science, Osaka University, Japan; Department of Protein Biochemistry, Institute of Life Science, Kurume University, Japan.
| |
Collapse
|
16
|
Lun CM, Waheed AA, Majadly A, Powell N, Freed EO. Mechanism of Viral Glycoprotein Targeting by Membrane-associated-RING-CH Proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.01.25.428025. [PMID: 33532773 PMCID: PMC7852266 DOI: 10.1101/2021.01.25.428025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
An emerging class of cellular inhibitory proteins has been identified that targets viral glycoproteins. These include the membrane-associated RING-CH (MARCH) family of E3 ubiquitin ligases that, among other functions, downregulate cell-surface proteins involved in adaptive immunity. The RING-CH domain of MARCH proteins is thought to function by catalyzing the ubiquitination of the cytoplasmic tails (CTs) of target proteins, leading to their degradation. MARCH proteins have recently been reported to target retroviral envelope glycoproteins (Env) and vesicular stomatitis virus G glycoprotein (VSV-G). However, the mechanism of antiviral activity remains poorly defined. Here we show that MARCH8 antagonizes the full-length forms of HIV-1 Env, VSV-G, Ebola virus glycoprotein (EboV-GP), and the spike (S) protein of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) thereby impairing the infectivity of virions pseudotyped with these viral glycoproteins. This MARCH8-mediated targeting of viral glycoproteins requires the E3 ubiquitin ligase activity of the RING-CH domain. We observe that MARCH8 protein antagonism of VSV-G is CT dependent. In contrast, MARCH8-mediated targeting of HIV-1 Env, EboV-GP and SARS-CoV-2 S protein by MARCH8 does not require the CT, suggesting a novel mechanism of MARCH-mediated antagonism of these viral glycoproteins. Confocal microscopy data demonstrate that MARCH8 traps the viral glycoproteins in an intracellular compartment. We observe that the endogenous expression of MARCH8 in several relevant human cell types is rapidly inducible by type I interferon. These results help to inform the mechanism by which MARCH proteins exert their antiviral activity and provide insights into the role of cellular inhibitory factors in antagonizing the biogenesis, trafficking, and virion incorporation of viral glycoproteins.
Collapse
Affiliation(s)
- Cheng Man Lun
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute
| | - Abdul A. Waheed
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute
| | - Alhlam Majadly
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute
| | - Nicole Powell
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute
| | - Eric O. Freed
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute
| |
Collapse
|
17
|
Chathuranga K, Kim TH, Lee H, Park JS, Kim JH, Chathuranga WAG, Ekanayaka P, Choi YJ, Lee CH, Kim CJ, Jung JU, Lee JS. Negative regulation of NEMO signaling by the ubiquitin E3 ligase MARCH2. EMBO J 2020; 39:e105139. [PMID: 32935379 PMCID: PMC7604578 DOI: 10.15252/embj.2020105139] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 01/01/2023] Open
Abstract
NF‐κB essential modulator (NEMO) is a key regulatory protein that functions during NF‐κB‐ and interferon‐mediated signaling in response to extracellular stimuli and pathogen infections. Tight regulation of NEMO is essential for host innate immune responses and for maintenance of homeostasis. Here, we report that the E3 ligase MARCH2 is a novel negative regulator of NEMO‐mediated signaling upon bacterial or viral infection. MARCH2 interacted directly with NEMO during the late phase of infection and catalyzed K‐48‐linked ubiquitination of Lys326 on NEMO, which resulted in its degradation. Deletion of MARCH2 resulted in marked resistance to bacterial/viral infection, along with increased innate immune responses both in vitro and in vivo. In addition, MARCH2−/− mice were more susceptible to LPS challenge due to massive production of cytokines. Taken together, these findings provide new insight into the molecular regulation of NEMO and suggest an important role for MARCH2 in homeostatic control of innate immune responses.
Collapse
Affiliation(s)
| | - Tae-Hwan Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Korea.,Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Hyuncheol Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Korea.,California Institute for Quantitative Biosciences, University of California, Berkeley, CA, USA
| | - Jun-Seol Park
- College of Veterinary Medicine, Chungnam National University, Daejeon, Korea
| | - Jae-Hoon Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology (UST), Daejeon, Korea
| | | | - Pathum Ekanayaka
- College of Veterinary Medicine, Chungnam National University, Daejeon, Korea
| | - Youn Jung Choi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology (UST), Daejeon, Korea
| | - Chul-Joong Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Korea
| | - Jae U Jung
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jong-Soo Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Korea
| |
Collapse
|
18
|
Jones E, Mead S. Genetic risk factors for Creutzfeldt-Jakob disease. Neurobiol Dis 2020; 142:104973. [PMID: 32565065 DOI: 10.1016/j.nbd.2020.104973] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/18/2020] [Accepted: 06/13/2020] [Indexed: 10/24/2022] Open
Abstract
Prion diseases are a group of fatal neurodegenerative disorders of mammals that share a central role for prion protein (PrP, gene PRNP) in their pathogenesis. Prions are infectious agents that account for the observed transmission of prion diseases between humans and animals in certain circumstances. The prion mechanism invokes a misfolded and multimeric assembly of PrP (a prion) that grows by templating of the normal protein and propagates by fission. Aside from the medical and public health notoriety of acquired prion diseases, the conditions have attracted interest as it has been realized that common neurodegenerative disorders share so-called prion-like mechanisms. In this article we will expand on recent evidence for new genetic loci that alter the risk of human prion disease. The most common human prion disease, sporadic Creutzfeldt-Jakob disease (sCJD), is characterized by the seemingly spontaneous appearance of prions in the brain. Genetic variation within PRNP is associated with all types of prion diseases, in particular, heterozygous genotypes at codons 129 and 219 have long been known to be strong protective factors against sCJD. A large number of rare mutations have been described in PRNP that cause autosomal dominant inherited prion diseases. Two loci recently identified by genome-wide association study increase sCJD risk, including variants in or near to STX6 and GAL3ST1. STX6 encodes syntaxin-6, a component of SNARE complexes with cellular roles that include the fusion of intracellular vesicles with target membranes. GAL3ST1 encodes cerebroside sulfotransferase, the only enzyme that sulfates sphingolipids to make sulfatides, a major lipid component of myelin. We discuss how these roles may modify the pathogenesis of prion diseases and their relevance for other neurodegenerative disorders.
Collapse
Affiliation(s)
- Emma Jones
- MRC Prion Unit at University College London (UCL), UCL Institute of Prion Diseases, 33 Cleveland Street, W1W 7FF, United Kingdom
| | - Simon Mead
- MRC Prion Unit at University College London (UCL), UCL Institute of Prion Diseases, 33 Cleveland Street, W1W 7FF, United Kingdom.
| |
Collapse
|
19
|
Lin H, Li S, Shu HB. The Membrane-Associated MARCH E3 Ligase Family: Emerging Roles in Immune Regulation. Front Immunol 2019; 10:1751. [PMID: 31404274 PMCID: PMC6669941 DOI: 10.3389/fimmu.2019.01751] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/10/2019] [Indexed: 01/13/2023] Open
Abstract
The membrane-associated RING-CH-type finger (MARCH) proteins of E3 ubiquitin ligases have emerged as critical regulators of immune responses. MARCH proteins target immune receptors, viral proteins as well as components in innate immune response for polyubiquitination and degradations via distinct routes. This review summarizes the current progress about MARCH proteins and their regulation on immune responses.
Collapse
Affiliation(s)
- Heng Lin
- Department of Infectious Diseases, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Shu Li
- Department of Infectious Diseases, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Hong-Bing Shu
- Department of Infectious Diseases, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
20
|
Yoo W, Cho EB, Kim S, Yoon JB. The E3 ubiquitin ligase MARCH2 regulates ERGIC3-dependent trafficking of secretory proteins. J Biol Chem 2019; 294:10900-10912. [PMID: 31142615 DOI: 10.1074/jbc.ra119.007435] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 05/17/2019] [Indexed: 11/06/2022] Open
Abstract
The E3 ubiquitin ligase membrane-associated ring-CH-type finger 2 (MARCH2) is known to be involved in intracellular vesicular trafficking, but its role in the early secretory pathway between the endoplasmic reticulum (ER) and Golgi compartments is largely unknown. Human ER-Golgi intermediate compartment protein 2 (ERGIC2) and ERGIC3 are orthologs of Erv41 and Erv46 in yeast, proteins that form a heteromeric complex, cycle between the ER and Golgi, and function as cargo receptors in both anterograde and retrograde protein trafficking. Here, we report that MARCH2 directs ubiquitination and subsequent degradation of ERGIC3 and that MARCH2 depletion increases endogenous ERGIC3 levels. We provide evidence that the lysine residues at positions 6 and 8 of ERGIC3 are the major sites of MARCH2-mediated ubiquitination. Of note, MARCH2 did not significantly decrease the levels of an ERGIC3 variant with lysine-to-arginine substitutions at residues 6 and 8. We also show that ERGIC3 binds to itself or to ERGIC2, whereas ERGIC2 is unable to interact with itself. Our results indicate that α1-antitrypsin and haptoglobin are likely to be cargo proteins of ERGIC3. We further observed that α1-antitrypsin and haptoglobin specifically bind to ERGIC3 and that ERGIC3 depletion decreases their secretion. Moreover, MARCH2 reduced secretion of α1-antitrypsin and haptoglobin, and coexpression of the ubiquitination-resistant ERGIC3 variant largely restored their secretion, suggesting that MARCH2-mediated ERGIC3 ubiquitination is the major cause of the decrease in trafficking of ERGIC3-binding secretory proteins. Our findings provide detailed insights into the regulation of the early secretory pathway by MARCH2 and into ERGIC3 function.
Collapse
Affiliation(s)
- Wonjin Yoo
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea and
| | - Eun-Bee Cho
- Department of Medical Lifesciences, Catholic University of Korea, Seoul 06591, Korea
| | - Sungjoo Kim
- Department of Medical Lifesciences, Catholic University of Korea, Seoul 06591, Korea
| | - Jong-Bok Yoon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea and.
| |
Collapse
|
21
|
Dingjan I, Linders PTA, Verboogen DRJ, Revelo NH, Ter Beest M, van den Bogaart G. Endosomal and Phagosomal SNAREs. Physiol Rev 2018; 98:1465-1492. [PMID: 29790818 DOI: 10.1152/physrev.00037.2017] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein family is of vital importance for organelle communication. The complexing of cognate SNARE members present in both the donor and target organellar membranes drives the membrane fusion required for intracellular transport. In the endocytic route, SNARE proteins mediate trafficking between endosomes and phagosomes with other endosomes, lysosomes, the Golgi apparatus, the plasma membrane, and the endoplasmic reticulum. The goal of this review is to provide an overview of the SNAREs involved in endosomal and phagosomal trafficking. Of the 38 SNAREs present in humans, 30 have been identified at endosomes and/or phagosomes. Many of these SNAREs are targeted by viruses and intracellular pathogens, which thereby reroute intracellular transport for gaining access to nutrients, preventing their degradation, and avoiding their detection by the immune system. A fascinating picture is emerging of a complex transport network with multiple SNAREs being involved in consecutive trafficking routes.
Collapse
Affiliation(s)
- Ilse Dingjan
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| | - Peter T A Linders
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| | - Danielle R J Verboogen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| | - Natalia H Revelo
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| | - Martin Ter Beest
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| | - Geert van den Bogaart
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| |
Collapse
|
22
|
Zhang Y, Lu J, Liu X. MARCH2 is upregulated in HIV-1 infection and inhibits HIV-1 production through envelope protein translocation or degradation. Virology 2018; 518:293-300. [PMID: 29573664 DOI: 10.1016/j.virol.2018.02.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 02/02/2018] [Accepted: 02/02/2018] [Indexed: 01/09/2023]
Abstract
MARCH2 is one of the MARCH family E3 ligases, which contains eleven members that play pivotal roles in controlling the turn-over of membrane proteins, such as MHC class I, MHC class II, and cell surface receptors. In this study, we found the expression of MARCH2 to be upregulated upon HIV-1 infection. MARCH2 inhibits the production and infection of HIV-1 through ligase activity-dependent envelope protein degradation and/or intracellular retention, a mechanism shared by MARCH8 that also leads to the inhibition of HIV-1 infection. Nevertheless, unlike MARCH8 and other MARCH proteins whose transcription levels are unrelated to viral infection, the expression level of MARCH2 is markedly upregulated upon HIV-1 infection, conferring MARCH2 a unique role in monitoring and regulating the HIV-1 infection-associated process.
Collapse
Affiliation(s)
- You Zhang
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jing Lu
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xinqi Liu
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
23
|
Yang X, Arines FM, Zhang W, Li M. Sorting of a multi-subunit ubiquitin ligase complex in the endolysosome system. eLife 2018; 7:33116. [PMID: 29355480 PMCID: PMC5811209 DOI: 10.7554/elife.33116] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 01/19/2018] [Indexed: 12/14/2022] Open
Abstract
The yeast Dsc E3 ligase complex has long been recognized as a Golgi-specific protein ubquitination system. It shares a striking sequence similarity to the Hrd1 complex that plays critical roles in the ER-associated degradation pathway. Using biochemical purification and mass spectrometry, we identified two novel Dsc subunits, which we named as Gld1 and Vld1. Surprisingly, Gld1 and Vld1 do not coexist in the same complex. Instead, they compete with each other to form two functionally independent Dsc subcomplexes. The Vld1 subcomplex takes the AP3 pathway to reach the vacuole membrane, whereas the Gld1 subcomplex travels through the VPS pathway and is cycled between Golgi and endosomes by the retromer. Thus, instead of being Golgi-specific, the Dsc complex can regulate protein levels at three distinct organelles, namely Golgi, endosome, and vacuole. Our study provides a novel model of achieving multi-tasking for transmembrane ubiquitin ligases with interchangeable trafficking adaptors. Proteins perform many tasks and, to remain healthy, each cell must ensure that its proteins are in good condition and present at the right levels. Plants, animals and fungi all largely deal with damaged, or otherwise unneeded, proteins by tagging them with a small marker called ubiquitin. The tagged proteins are then rapidly destroyed, which prevents them from harming the cells. Enzymes known as E3 ligases attach ubiquitin to proteins. Yet, the number of E3 ligases is dwarfed by the number of proteins modified with ubiquitin. For instance, humans have approximately 20,000 different proteins, about one third of which are found in or on cell membranes. However, there are only around 600 E3 ligases, and only about 50 of them are associated with cell membranes. This is further complicated by the fact that proteins are also present in distinct compartments within the cell. The Dsc complex, for example, is an E3 ligase from yeast that is found within a compartment of the cell known as the Golgi. It was thus expected to only attach ubiquitin to Golgi proteins. Yet some recent studies showed that the Dsc complex could also tag proteins present in two other compartments of yeast cells: the endosome and vacuole. How can the Dsc complex act on proteins in three distinct compartments? The Dsc complex is actually made from multiple proteins, and Yang et al. now report two new protein components. Biochemical and genetic tools showed that these two proteins do not co-exist in the same Dsc complex. Instead, they compete with each other to form two different kinds of Dsc complexes, which Yang et al. refer to as subcomplexes. Further work showed that the two new proteins determine the route taken by the Dsc complex along the cell’s protein transport pathway. One subcomplex is transported to the vacuole and the other cycles between the Golgi and endosomes. Thus, by changing just one component, the Dsc complex can be sent to different locations within the cell. These findings describe a new mechanism that enables E3 ligases to multi-task on a wide range of proteins, even across distinct compartments of the cell. Future work will determine whether plant and animal cells also use a similar strategy. Since defects in protein quality control contribute to many human diseases, such as Alzheimer's and Parkinson's disease, working out how E3 ligases work is important for the field of biomedicine.
Collapse
Affiliation(s)
- Xi Yang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Felichi Mae Arines
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Weichao Zhang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Ming Li
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, United States
| |
Collapse
|
24
|
Lee H, Cheong SM, Han W, Koo Y, Jo SB, Cho GS, Yang JS, Kim S, Han JK. March2 is required for head formation by mediating Dishevelled degradation in concert with Dapper1. Development 2018; 145:dev.143107. [DOI: 10.1242/dev.143107] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 02/21/2018] [Indexed: 11/20/2022]
Abstract
Dishevelled (Dvl/Dsh) is a key scaffold protein that propagates Wnt signaling essential for embryogenesis and homeostasis. However, whether antagonism of Wnt signaling necessary for vertebrate head formation can be achieved through regulation of Dsh protein stability is unclear. Here we show that membrane-associated RING-CH2 (March2), a RING-type E3 ubiquitin ligase, antagonizes Wnt signaling by regulating the turnover of Dsh protein via ubiquitin-mediated lysosomal degradation in prospective head region of Xenopus. We further found that March2 acquires regional and functional specificities for head formation from the Dsh-interacting protein Dapper1 (Dpr1). Dpr1 stabilizes interaction between March2 and Dsh for mediating ubiquitination and subsequent degradation of Dsh protein only in the dorso-animal region of Xenopus embryo. These results suggest that March2 restricts cytosolic pools of Dsh protein and leads to subsequent limitation of Wnt signaling for the precise vertebrate head development.
Collapse
Affiliation(s)
- Hyeyoon Lee
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea
- Present address: Division of Molecular Embryology, DKFZ-ZMBH Alliance, Deutsches Krebsforschungszentrum (DKFZ), 69120 Heidelberg, Germany
| | - Seong-Moon Cheong
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea
- Present address: Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wonhee Han
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea
| | - Youngmu Koo
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea
| | - Saet-Byeol Jo
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea
| | - Gun-Sik Cho
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea
- Present address: Laboratory of Stem Cells, NEXEL, Seoul, Republic of Korea
| | - Jae-Seong Yang
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea
- Present address: EMBL/CRG Systems Biology Research Unit, Center for Genomic Regulation, Dr. Aiguader 88, Barcelona, Spain
| | - Sanguk Kim
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea
| | - Jin-Kwan Han
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea
| |
Collapse
|
25
|
Bauer J, Bakke O, Morth JP. Overview of the membrane-associated RING-CH (MARCH) E3 ligase family. N Biotechnol 2017; 38:7-15. [DOI: 10.1016/j.nbt.2016.12.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/05/2016] [Accepted: 12/13/2016] [Indexed: 12/17/2022]
|
26
|
Knockout of MARCH2 inhibits the growth of HCT116 colon cancer cells by inducing endoplasmic reticulum stress. Cell Death Dis 2017; 8:e2957. [PMID: 28749466 PMCID: PMC5584615 DOI: 10.1038/cddis.2017.347] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 06/16/2017] [Accepted: 06/19/2017] [Indexed: 02/06/2023]
Abstract
Membrane-associated RING-CH protein 2 (MARCH2), a member of the MARCH family, functions in vesicle trafficking and autophagy regulation. In this study, we established MARCH2 knockout HCT116 cell lines using CRISPR/Cas9-mediated genome editing to evaluate the role of MARCH2 in colon cancer in vitro and in vivo. Knockout of MARCH2 suppressed cell proliferation, and promoted autophagy, apoptosis and G2/M phase cell cycle arrest. These effects were associated with activation of endoplasmic reticulum (ER) stress. In addition, loss of MARCH2 sensitized HCT116 cells to the chemotherapy drugs etoposide and cisplatin. Moreover, we analyzed the clinical significance of MARCH2 in human colon carcinoma (n=100). High MARCH2 expression was significantly associated with advanced clinicopathological features and poorer overall survival in colon carcinoma. MARCH2 expression correlated negatively with expression of the unfolded protein response molecule p-PERK in colon cancer. Collectively, these data reveal a relationship between MARCH2, ER stress and colon cancer, and indicates MARCH2 may have an important role in the development and progression of colon cancer.
Collapse
|
27
|
Abstract
Newly synthesized transmembrane proteins undergo a series of steps to ensure that only the required amount of correctly folded protein is localized to the membrane. The regulation of protein quality and its abundance at the membrane are often controlled by ubiquitination, a multistep enzymatic process that results in the attachment of ubiquitin, or chains of ubiquitin to the target protein. Protein ubiquitination acts as a signal for sorting, trafficking, and the removal of membrane proteins via endocytosis, a process through which multiple ubiquitin ligases are known to specifically regulate the functions of a number of ion channels, transporters, and signaling receptors. Endocytic removal of these proteins through ubiquitin-dependent endocytosis provides a way to rapidly downregulate the physiological outcomes, and defects in such controls are directly linked to human pathologies. Recent evidence suggests that ubiquitination is also involved in the shedding of membranes and associated proteins as extracellular vesicles, thereby not only controlling the cell surface levels of some membrane proteins, but also their potential transport to neighboring cells. In this review, we summarize the mechanisms and functions of ubiquitination of membrane proteins and provide specific examples of ubiquitin-dependent regulation of membrane proteins.
Collapse
Affiliation(s)
- Natalie Foot
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Tanya Henshall
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| |
Collapse
|
28
|
USP19-Mediated Deubiquitination Facilitates the Stabilization of HRD1 Ubiquitin Ligase. Int J Mol Sci 2016; 17:ijms17111829. [PMID: 27827840 PMCID: PMC5133830 DOI: 10.3390/ijms17111829] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/26/2016] [Accepted: 10/27/2016] [Indexed: 01/28/2023] Open
Abstract
In the endoplasmic reticulum (ER), misfolded and unfolded proteins are eliminated by a process called ER-associated protein degradation (ERAD) in order to maintain cell homeostasis. In the ERAD pathway, several ER-localized E3 ubiquitin ligases target ERAD substrate proteins for ubiquitination and subsequent proteasomal degradation. However, little is known about how the functions of the ERAD ubiquitin ligases are regulated. Recently, USP19, an ER-anchored deubiquitinating enzyme (DUB), has been suggested to be involved in the regulation of ERAD. In this study, HRD1, an ERAD ubiquitin ligase, is shown to be a novel substrate for USP19. We demonstrate that USP19 rescues HRD1 from proteasomal degradation by deubiquitination of K48-linked ubiquitin chains. In addition, the altered expression of USP19 affects the steady-state levels of HRD1. These results suggest that USP19 regulates the stability of HRD1 and provide insight into the regulatory mechanism of the ERAD ubiquitin ligases.
Collapse
|
29
|
Xia D, Qu L, Li G, Hongdu B, Xu C, Lin X, Lou Y, He Q, Ma D, Chen Y. MARCH2 regulates autophagy by promoting CFTR ubiquitination and degradation and PIK3CA-AKT-MTOR signaling. Autophagy 2016; 12:1614-30. [PMID: 27308891 DOI: 10.1080/15548627.2016.1192752] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
MARCH2 (membrane-associated RING-CH protein 2), an E3 ubiquitin ligase, is mainly associated with the vesicle trafficking. In the present study, for the first time, we demonstrated that MARCH2 negatively regulates autophagy. Our data indicated that overexpression of MARCH2 impaired autophagy, as evidenced by attenuated levels of LC3B-II and impaired degradation of endogenous and exogenous autophagic substrates. By contrast, loss of MARCH2 expression had the opposite effects. In vivo experiments demonstrate that MARCH2 knockout mediated autophagy results in an inhibition of tumorigenicity. Further investigation revealed that the induction of autophagy by MARCH2 deficiency was mediated through the PIK3CA-AKT-MTOR signaling pathway. Additionally, we found that MARCH2 interacts with CFTR (cystic fibrosis transmembrane conductance regulator), promotes the ubiquitination and degradation of CFTR, and inhibits CFTR-mediated autophagy in tumor cells. The functional PDZ domain of MARCH2 is required for the association with CFTR. Thus, our study identified a novel negative regulator of autophagy and suggested that the physical and functional connection between the MARCH2 and CFTR in different conditions will be elucidated in the further experiments.
Collapse
Affiliation(s)
- Dan Xia
- a Department of Immunology , Peking University School of Basic Medical Sciences , Beijing , China.,b Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Sciences Center , Beijing , China.,c Center for Human Disease Genomics, Peking University , Beijing , China
| | - Liujing Qu
- a Department of Immunology , Peking University School of Basic Medical Sciences , Beijing , China.,b Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Sciences Center , Beijing , China.,c Center for Human Disease Genomics, Peking University , Beijing , China
| | - Ge Li
- a Department of Immunology , Peking University School of Basic Medical Sciences , Beijing , China.,b Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Sciences Center , Beijing , China.,c Center for Human Disease Genomics, Peking University , Beijing , China
| | - Beiqi Hongdu
- a Department of Immunology , Peking University School of Basic Medical Sciences , Beijing , China.,b Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Sciences Center , Beijing , China.,c Center for Human Disease Genomics, Peking University , Beijing , China
| | - Chentong Xu
- a Department of Immunology , Peking University School of Basic Medical Sciences , Beijing , China.,b Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Sciences Center , Beijing , China.,c Center for Human Disease Genomics, Peking University , Beijing , China
| | - Xin Lin
- a Department of Immunology , Peking University School of Basic Medical Sciences , Beijing , China.,b Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Sciences Center , Beijing , China.,c Center for Human Disease Genomics, Peking University , Beijing , China
| | - Yaxin Lou
- d Medical and Healthy Analytical Center, Peking University , Beijing , China
| | - Qihua He
- d Medical and Healthy Analytical Center, Peking University , Beijing , China
| | - Dalong Ma
- a Department of Immunology , Peking University School of Basic Medical Sciences , Beijing , China.,b Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Sciences Center , Beijing , China.,c Center for Human Disease Genomics, Peking University , Beijing , China
| | - Yingyu Chen
- a Department of Immunology , Peking University School of Basic Medical Sciences , Beijing , China.,b Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Sciences Center , Beijing , China.,c Center for Human Disease Genomics, Peking University , Beijing , China
| |
Collapse
|
30
|
Jean-Charles PY, Snyder JC, Shenoy SK. Chapter One - Ubiquitination and Deubiquitination of G Protein-Coupled Receptors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 141:1-55. [PMID: 27378754 DOI: 10.1016/bs.pmbts.2016.05.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The seven-transmembrane containing G protein-coupled receptors (GPCRs) constitute the largest family of cell-surface receptors. Transmembrane signaling by GPCRs is fundamental to many aspects of physiology including vision, olfaction, cardiovascular, and reproductive functions as well as pain, behavior and psychomotor responses. The duration and magnitude of signal transduction is tightly controlled by a series of coordinated trafficking events that regulate the cell-surface expression of GPCRs at the plasma membrane. Moreover, the intracellular trafficking profiles of GPCRs can correlate with the signaling efficacy and efficiency triggered by the extracellular stimuli that activate GPCRs. Of the various molecular mechanisms that impart selectivity, sensitivity and strength of transmembrane signaling, ubiquitination of the receptor protein plays an important role because it defines both trafficking and signaling properties of the activated GPCR. Ubiquitination of proteins was originally discovered in the context of lysosome-independent degradation of cytosolic proteins by the 26S proteasome; however a large body of work suggests that ubiquitination also orchestrates the downregulation of membrane proteins in the lysosomes. In the case of GPCRs, such ubiquitin-mediated lysosomal degradation engenders long-term desensitization of transmembrane signaling. To date about 40 GPCRs are known to be ubiquitinated. For many GPCRs, ubiquitination plays a major role in postendocytic trafficking and sorting to the lysosomes. This chapter will focus on the patterns and functional roles of GPCR ubiquitination, and will describe various molecular mechanisms involved in GPCR ubiquitination.
Collapse
Affiliation(s)
- P-Y Jean-Charles
- Department of Medicine (Cardiology), Duke University Medical Center, Durham, NC, United States
| | - J C Snyder
- Department of Cell Biology, Duke University Medical Center, Durham, NC, United States
| | - S K Shenoy
- Department of Medicine (Cardiology), Duke University Medical Center, Durham, NC, United States; Department of Cell Biology, Duke University Medical Center, Durham, NC, United States.
| |
Collapse
|
31
|
Grassi D, Plonka FB, Oksdath M, Guil AN, Sosa LJ, Quiroga S. Selected SNARE proteins are essential for the polarized membrane insertion of igf-1 receptor and the regulation of initial axonal outgrowth in neurons. Cell Discov 2015; 1:15023. [PMID: 27462422 PMCID: PMC4860833 DOI: 10.1038/celldisc.2015.23] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 07/07/2015] [Indexed: 02/08/2023] Open
Abstract
The establishment of polarity necessitates initial axonal outgrowth and,
therefore, the addition of new membrane to the axon’s plasmalemma.
Axolemmal expansion occurs by exocytosis of plasmalemmal precursor vesicles
(PPVs) primarily at the neuronal growth cone. Little is known about the SNAREs
family proteins involved in the regulation of PPV fusion with the neuronal
plasmalemma at early stages of differentiation. We show here that five SNARE
proteins (VAMP2, VAMP4, VAMP7, Syntaxin6 and SNAP23) were expressed by
hippocampal pyramidal neurons before polarization. Expression silencing of three
of these proteins (VAMP4, Syntaxin6 and SNAP23) repressed axonal outgrowth and
the establishment of neuronal polarity, by inhibiting IGF-1 receptor exocytotic
polarized insertion, necessary for neuronal polarization. In addition,
stimulation with IGF-1 triggered the association of VAMP4, Syntaxin6 and SNAP23
to vesicular structures carrying the IGF-1 receptor and overexpression of a
negative dominant form of Syntaxin6 significantly inhibited exocytosis of IGF-1
receptor containing vesicles at the neuronal growth cone. Taken together, our
results indicated that VAMP4, Syntaxin6 and SNAP23 functions are essential for
regulation of PPV exocytosis and the polarized insertion of IGF-1 receptor and,
therefore, required for initial axonal elongation and the establishment of
neuronal polarity.
Collapse
Affiliation(s)
- Diego Grassi
- Departamento de Química Biológica-CIQUIBIC, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba-CONICET , Córdoba, Argentina
| | - Florentyna Bustos Plonka
- Departamento de Química Biológica-CIQUIBIC, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba-CONICET , Córdoba, Argentina
| | - Mariana Oksdath
- Departamento de Química Biológica-CIQUIBIC, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba-CONICET , Córdoba, Argentina
| | - Alvaro Nieto Guil
- Departamento de Química Biológica-CIQUIBIC, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba-CONICET , Córdoba, Argentina
| | - Lucas J Sosa
- Departamento de Química Biológica-CIQUIBIC, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba-CONICET , Córdoba, Argentina
| | - Santiago Quiroga
- Departamento de Química Biológica-CIQUIBIC, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba-CONICET , Córdoba, Argentina
| |
Collapse
|
32
|
Souza Melo CP, Campos CB, Dutra ÁP, Neto JCA, Fenelon AJS, Neto AH, Carbone EK, Pianovski MAD, Ferreira ACDS, Assumpcão JG. Correlation between FLT3-ITD status and clinical, cellular and molecular profiles in promyelocytic acute leukemias. Leuk Res 2014; 39:131-7. [PMID: 25530565 DOI: 10.1016/j.leukres.2014.11.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 11/11/2014] [Accepted: 11/20/2014] [Indexed: 10/24/2022]
Abstract
Internal tandem duplications (ITD) of FLT3 gene occur in about a third of acute promyelocytic leukemias (APL). We investigated the patterns of blood count, surface antigen, expression, chromosome aberrations, PML-RARa isoform, gene expression profile (GEP) and survival in 34 APL patients according to FLT3-ITD status. 97% had a t(15;17) and all of them carried PML-RARa gene fusion, 8 (23.5%) had a FLT3-ITD mutation. Presence of ITD was associated with higher Hb and WBC levels, bcr3 isoform, CD34 expression, CD2 or CD2/CD34 expression. In a multivariate analysis, Hb>9.6g/dL and WBC≥20 × 10(9)/L were important factors for predicting ITD presence. GEP showed that FLT3-ITD carriers clustered separately, even when as few as 5 genes were considered. This study provides further evidence that FLT3-ITDs carriers constitute a biologically distinct group of APL patients.
Collapse
Affiliation(s)
- Carolina Pereira Souza Melo
- Laboratório BIOCOD Biotecnologia Ltda., Av. das Nações 2448, Portaria A, Vespasiano CEP 33200-000, MG, Brazil.
| | - Catharina Brant Campos
- Laboratório BIOCOD Biotecnologia Ltda., Av. das Nações 2448, Portaria A, Vespasiano CEP 33200-000, MG, Brazil.
| | - Álvaro Pimenta Dutra
- Hospital Santa Casa de Misericórdia de Belo Horizonte, Av. Francisco Sales 1111, Belo Horizonte CEP 30150-221, MG, Brazil.
| | - Joaquim Caetano Aguirre Neto
- Hospital Santa Casa de Misericórdia de Belo Horizonte, Av. Francisco Sales 1111, Belo Horizonte CEP 30150-221, MG, Brazil.
| | - Alexandre José Silva Fenelon
- Hospital Santa Casa de Misericórdia de Belo Horizonte, Av. Francisco Sales 1111, Belo Horizonte CEP 30150-221, MG, Brazil.
| | - Abrahão Hallack Neto
- Hospital Universitário da Universidade Federal de Juiz de Fora, Rua Catulo Breviglieri s/n, Juiz de Fora CEP 36036-110, MG, Brazil.
| | - Edna Kakitani Carbone
- Hospital Pequeno Príncipe, Rua Desembargador Motta 1070, Curitiba CEP 80250-060, PR, Brazil.
| | | | | | - Juliana Godoy Assumpcão
- Laboratório BIOCOD Biotecnologia Ltda., Av. das Nações 2448, Portaria A, Vespasiano CEP 33200-000, MG, Brazil; Setor de Pesquisa e Desenvolvimento-Instituto Hermes Pardini, Av. das Nações 2448, Portaria A, Vespasiano CEP 33200-000, MG, Brazil.
| |
Collapse
|
33
|
The Membrane Associated RING-CH Proteins: A Family of E3 Ligases with Diverse Roles through the Cell. INTERNATIONAL SCHOLARLY RESEARCH NOTICES 2014; 2014:637295. [PMID: 27419207 PMCID: PMC4897099 DOI: 10.1155/2014/637295] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 08/22/2014] [Indexed: 01/03/2023]
Abstract
Since the discovery that conjugation of ubiquitin to proteins can drive proteolytic degradation, ubiquitination has been shown to perform a diverse range of functions in the cell. It plays an important role in endocytosis, signal transduction, trafficking of vesicles inside the cell, and even DNA repair. The process of ubiquitination-mediated control has turned out to be remarkably complex, involving a diverse array of proteins and many levels of control. This review focuses on a family of structurally related E3 ligases termed the membrane-associated RING-CH (MARCH) ubiquitin ligases, which were originally discovered as structural homologs to the virals E3s, K3, and K5 from Kaposi's sarcoma-associated herpesvirus (KSHV). These proteins contain a catalytic RING-CH finger and are typically membrane-bound, with some having up to 14 putative transmembrane domains. Despite several lines of evidence showing that the MARCH proteins play a complex and essential role in several cellular processes, this family remains understudied.
Collapse
|
34
|
Funakoshi Y, Chou MM, Kanaho Y, Donaldson JG. TRE17/USP6 regulates ubiquitylation and trafficking of cargo proteins that enter cells by clathrin-independent endocytosis. J Cell Sci 2014; 127:4750-61. [PMID: 25179595 DOI: 10.1242/jcs.156786] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Plasma membrane proteins that enter cells by clathrin-independent endocytosis (CIE) are sorted either to lysosomes for degradation or recycled back to the plasma membrane. Expression of some MARCH E3 ubiquitin ligases promotes trafficking of CIE cargo proteins to lysosomes by ubiquitylating the proteins. Here, we show that co-expression of the ubiquitin-specific protease TRE17/USP6 counteracts the MARCH-dependent targeting of CIE cargo proteins, but not that of transferrin receptor, to lysosomes, leading to recovery of the stability and cell surface level of the proteins. The ubiquitylation of CIE cargo proteins by MARCH8 was reversed by TRE17, suggesting that TRE17 leads to deubiquitylation of CIE cargo proteins. The effects of TRE17 were dependent on its deubiquitylating activity and expression of TRE17 alone led to a stabilization of surface major histocompatibility complex class I (MHCI) molecules, a CIE cargo, suggesting that deubiquitylation of endogenous CIE cargo proteins promotes their stability. This study demonstrates that cycles of ubiquitylation and deubiquitylation can determine whether CIE cargo proteins are degraded or recycled.
Collapse
Affiliation(s)
- Yuji Funakoshi
- Cell Biology & Physiology Center, NHLBI, NIH, Bethesda, MD 20891, USA Department of Physiological Chemistry, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Ten-nodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Margaret M Chou
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, 3615 Civic Center Boulevard, Philadelphia, PA19104, USA
| | - Yasunori Kanaho
- Department of Physiological Chemistry, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Ten-nodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Julie G Donaldson
- Cell Biology & Physiology Center, NHLBI, NIH, Bethesda, MD 20891, USA
| |
Collapse
|
35
|
Sirisaengtaksin N, Gireud M, Yan Q, Kubota Y, Meza D, Waymire JC, Zage PE, Bean AJ. UBE4B protein couples ubiquitination and sorting machineries to enable epidermal growth factor receptor (EGFR) degradation. J Biol Chem 2013; 289:3026-39. [PMID: 24344129 DOI: 10.1074/jbc.m113.495671] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The signaling of plasma membrane proteins is tuned by internalization and sorting in the endocytic pathway prior to recycling or degradation in lysosomes. Ubiquitin modification allows recognition and association of cargo with endosomally associated protein complexes, enabling sorting of proteins to be degraded from those to be recycled. The mechanism that provides coordination between the cellular machineries that mediate ubiquitination and endosomal sorting is unknown. We report that the ubiquitin ligase UBE4B is recruited to endosomes in response to epidermal growth factor receptor (EGFR) activation by binding to Hrs, a key component of endosomal sorting complex required for transport (ESCRT) 0. We identify the EGFR as a substrate for UBE4B, establish UBE4B as a regulator of EGFR degradation, and describe a mechanism by which UBE4B regulates endosomal sorting, affecting cellular levels of the EGFR and its downstream signaling. We propose a model in which the coordinated action of UBE4B, ESCRT-0, and the deubiquitinating enzyme USP8 enable the endosomal sorting and lysosomal degradation of the EGFR.
Collapse
Affiliation(s)
- Natalie Sirisaengtaksin
- From the Department of Neurobiology and Anatomy and the Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas 77030
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Cheng J, Guggino W. Ubiquitination and degradation of CFTR by the E3 ubiquitin ligase MARCH2 through its association with adaptor proteins CAL and STX6. PLoS One 2013; 8:e68001. [PMID: 23818989 PMCID: PMC3688601 DOI: 10.1371/journal.pone.0068001] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 05/29/2013] [Indexed: 12/23/2022] Open
Abstract
Golgi-localized cystic fibrosis transmembrane conductance regulator (CFTR)-associated ligand (CAL) and syntaxin 6 (STX6) regulate the abundance of mature, post-ER CFTR by forming a CAL/STX6/CFTR complex (CAL complex) that promotes CFTR degradation in lysosomes. However, the molecular mechanism underlying this degradation is unknown. Here we investigated the interaction of a Golgi-localized, membrane-associated RING-CH E3 ubiquitin ligase, MARCH2, with the CAL complex and the consequent binding, ubiquitination, and degradation of mature CFTR. We found that MARCH2 not only co-immunoprecipitated and co-localized with CAL and STX6, but its binding to CAL was also enhanced by STX6, suggesting a synergistic interaction. In vivo ubiquitination assays demonstrated the ubiquitination of CFTR by MARCH2, and overexpression of MARCH2, like that of CAL and STX6, led to a dose-dependent degradation of mature CFTR that was blocked by bafilomycin A1 treatment. A catalytically dead MARCH2 RING mutant was unable to promote CFTR degradation. In addition, MARCH2 had no effect on a CFTR mutant lacking the PDZ motif, suggesting that binding to the PDZ domain of CAL is required for MARCH2-mediated degradation of CFTR. Indeed, silencing of endogenous CAL ablated the effect of MARCH2 on CFTR. Consistent with its Golgi localization, MARCH2 had no effect on ER-localized ΔF508-CFTR. Finally, siRNA-mediated silencing of endogenous MARCH2 in the CF epithelial cell line CFBE-CFTR increased the abundance of mature CFTR. Taken together, these data suggest that the recruitment of the E3 ubiquitin ligase MARCH2 to the CAL complex and subsequent ubiquitination of CFTR are responsible for the CAL-mediated lysosomal degradation of mature CFTR.
Collapse
Affiliation(s)
- Jie Cheng
- Department of Physiology, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
| | - William Guggino
- Department of Physiology, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
37
|
Holloway ZG, Velayos-Baeza A, Howell GJ, Levecque C, Ponnambalam S, Sztul E, Monaco AP. Trafficking of the Menkes copper transporter ATP7A is regulated by clathrin-, AP-2-, AP-1-, and Rab22-dependent steps. Mol Biol Cell 2013; 24:1735-48, S1-8. [PMID: 23596324 PMCID: PMC3667726 DOI: 10.1091/mbc.e12-08-0625] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
ATP7A mediates copper absorption and feeds cuproenzymes in the trans-Golgi network. To regulate copper homeostasis, ATP7A cycles between the TGN and plasma membrane. The roles of clathrin, adaptor complexes, lipid rafts, and Rab22a are assessed in an attempt to decipher the regulatory proteins involved in ATP7A cycling. The transporter ATP7A mediates systemic copper absorption and provides cuproenzymes in the trans-Golgi network (TGN) with copper. To regulate metal homeostasis, ATP7A constitutively cycles between the TGN and plasma membrane (PM). ATP7A trafficking to the PM is elevated in response to increased copper load and is reversed when copper concentrations are lowered. Molecular mechanisms underlying this trafficking are poorly understood. We assess the role of clathrin, adaptor complexes, lipid rafts, and Rab22a in an attempt to decipher the regulatory proteins involved in ATP7A cycling. While RNA interference (RNAi)–mediated depletion of caveolin 1/2 or flotillin had no effect on ATP7A localization, clathrin heavy chain depletion or expression of AP180 dominant-negative mutant not only disrupted clathrin-regulated pathways, but also blocked PM-to-TGN internalization of ATP7A. Depletion of the μ subunits of either adaptor protein-2 (AP-2) or AP-1 using RNAi further provides evidence that both clathrin adaptors are important for trafficking of ATP7A from the PM to the TGN. Expression of the GTP-locked Rab22aQ64L mutant caused fragmentation of TGN membrane domains enriched for ATP7A. These appear to be a subdomain of the mammalian TGN, showing only partial overlap with the TGN marker golgin-97. Of importance, ATP7A remained in the Rab22aQ64L-generated structures after copper treatment and washout, suggesting that forward trafficking out of this compartment was blocked. This study provides evidence that multiple membrane-associated factors, including clathrin, AP-2, AP-1, and Rab22, are regulators of ATP7A trafficking.
Collapse
Affiliation(s)
- Zoe G Holloway
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
38
|
Han SO, Xiao K, Kim J, Wu JH, Wisler JW, Nakamura N, Freedman NJ, Shenoy SK. MARCH2 promotes endocytosis and lysosomal sorting of carvedilol-bound β(2)-adrenergic receptors. ACTA ACUST UNITED AC 2012; 199:817-30. [PMID: 23166351 PMCID: PMC3514787 DOI: 10.1083/jcb.201208192] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The β2-adrenergic receptor antagonist carvedilol recruits MARCH2, a unique E3 ubiquitin ligase, to promote receptor endocytosis and lysosomal trafficking. Lysosomal degradation of ubiquitinated β2-adrenergic receptors (β2ARs) serves as a major mechanism of long-term desensitization in response to prolonged agonist stimulation. Surprisingly, the βAR antagonist carvedilol also induced ubiquitination and lysosomal trafficking of both endogenously expressed β2ARs in vascular smooth muscle cells (VSMCs) and overexpressed Flag-β2ARs in HEK-293 cells. Carvedilol prevented β2AR recycling, blocked recruitment of Nedd4 E3 ligase, and promoted the dissociation of the deubiquitinases USP20 and USP33. Using proteomics approaches (liquid chromatography–tandem mass spectrometry), we identified that the E3 ligase MARCH2 interacted with carvedilol-bound β2AR. The association of MARCH2 with internalized β2ARs was stabilized by carvedilol and did not involve β-arrestin. Small interfering RNA–mediated down-regulation of MARCH2 ablated carvedilol-induced ubiquitination, endocytosis, and degradation of endogenous β2ARs in VSMCs. These findings strongly suggest that specific ligands recruit distinct E3 ligase machineries to activated cell surface receptors and direct their intracellular itinerary. In response to β blocker therapy with carvedilol, MARCH2 E3 ligase activity regulates cell surface β2AR expression and, consequently, its signaling.
Collapse
Affiliation(s)
- Sang-oh Han
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Intracellular membrane trafficking along endocytic and secretory transport pathways plays a critical role in diverse cellular functions including both developmental and pathological processes. Briefly, proteins and lipids destined for transport to distinct locations are collectively assembled into vesicles and delivered to their target site by vesicular fusion. SNARE (soluble N-ethylmaleimide-sensitive factor-attachment protein receptor) proteins are required for these events, during which v-SNAREs (vesicle SNAREs) interact with t-SNAREs (target SNAREs) to allow transfer of cargo from donor vesicle to target membrane. Recently, the t-SNARE family member, syntaxin-6, has been shown to play an important role in the transport of proteins that are key to diverse cellular dynamic processes. In this paper, we briefly discuss the specific role of SNAREs in various mammalian cell types and comprehensively review the various roles of the Golgi- and endosome-localized t-SNARE, syntaxin-6, in membrane trafficking during physiological as well as pathological conditions.
Collapse
|
40
|
Comparison of splenic transcriptome activity of two rainbow trout strains differing in robustness under regional aquaculture conditions. Mol Biol Rep 2012; 40:1955-66. [DOI: 10.1007/s11033-012-2252-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 10/10/2012] [Indexed: 10/27/2022]
|
41
|
Bartnikas TB, Parker CC, Cheng R, Campagna DR, Lim JE, Palmer AA, Fleming MD. QTLs for murine red blood cell parameters in LG/J and SM/J F(2) and advanced intercross lines. Mamm Genome 2012; 23:356-66. [PMID: 22322356 PMCID: PMC3358495 DOI: 10.1007/s00335-012-9393-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 01/11/2012] [Indexed: 10/14/2022]
Abstract
Red blood cells are essential for oxygen transport and other physiologic processes. Red cell characteristics are typically determined by complete blood counts which measure parameters such as hemoglobin levels and mean corpuscular volumes; these parameters reflect the quality and quantity of red cells in the circulation at any particular moment. To identify the genetic determinants of red cell parameters, we performed genome-wide association analysis on LG/J×SM/J F2 and F34 advanced intercross lines using single nucleotide polymorphism genotyping and a novel algorithm for mapping in the combined populations. We identified significant quantitative trait loci for red cell parameters on chromosomes 6, 7, 8, 10, 12, and 17; our use of advanced intercross lines reduced the quantitative trait loci interval width from 1.6- to 9.4-fold. Using the genomic sequences of LG/J and SM/J mice, we identified nonsynonymous coding single nucleotide polymorphisms in candidate genes residing within quantitative trait loci and performed sequence alignments and molecular modeling to gauge the potential impact of amino acid substitutions. These results should aid in the identification of genes critical for red cell physiology and metabolism and demonstrate the utility of advanced intercross lines in uncovering genetic determinants of inherited traits.
Collapse
Affiliation(s)
- Thomas B Bartnikas
- Department of Pathology, Children's Hospital, Enders 1110, 300 Longwood Avenue, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Hassink G, Slotman J, Oorschot V, Van Der Reijden BA, Monteferrario D, Noordermeer SM, Van Kerkhof P, Klumperman J, Strous GJ. Identification of the ubiquitin ligase Triad1 as a regulator of endosomal transport. Biol Open 2012; 1:607-14. [PMID: 23213454 PMCID: PMC3509441 DOI: 10.1242/bio.2012778] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The ubiquitin system plays an important role in trafficking of signaling receptors from the plasma membrane to lysosomes. Triad1 is a ubiquitin ligase that catalyzes the formation of poly-ubiquitin chains linked via lysine-48 as well as lysine-63 residues. We show that depletion of Triad1 affects the sorting of both growth hormone and epidermal growth factor. Triad1-depleted cells accumulate both ligands in endosomes. While fluid phase transport to the lysosomes is reduced in the absence of Triad1, growth hormone receptor can recycle back to the plasma membrane together with transferrin. Using immune electron microscopy we show that Triad1 depletion results in enlarged endosomes with enlarged and irregular shaped intraluminal vesicles. The endosomes display prominent clathrin coats and show increased levels of growth hormone label. We conclude that Triad1 is required for the proper function of multivesicular bodies.
Collapse
Affiliation(s)
- Gerco Hassink
- Department of Cell Biology and Institute of Biomembranes, University Medical Center Utrecht , 3584 CX Utrecht , The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
MacDonald JIS, Dietrich A, Gamble S, Hryciw T, Grant RI, Meakin SO. Nesca, a novel neuronal adapter protein, links the molecular motor kinesin with the pre-synaptic membrane protein, syntaxin-1, in hippocampal neurons. J Neurochem 2012; 121:861-80. [PMID: 22404429 DOI: 10.1111/j.1471-4159.2012.07729.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Vesicular transport in neurons plays a vital role in neuronal function and survival. Nesca is a novel protein that we previously identified and herein describe its pattern of expression, subcellular localization and protein-protein interactions both in vitro and in vivo. Specifically, a large proportion of Nesca is in tight association with both actin and microtubule cytoskeletal proteins. Nesca binds to F-actin, microtubules, βIII and acetylated α-tubulin, but not neurofilaments or the actin-binding protein drebrin, in in vitro-binding assays. Nesca co-immunoprecipitates with kinesin heavy chain (KIF5B) and kinesin light-chain motors as well as with the synaptic membrane precursor protein, syntaxin-1, and is a constituent of the post-synaptic density. Moreover, in vitro-binding assays indicate that Nesca directly binds KIF5B, kinesin light-chain and syntaxin-1. In contrast, Nesca does not co-immunoprecipitate with the kinesin motors KIF1B, KIF3A nor does it bind syntaxin-4 or the synaptosome-associated protein 25 kDa (SNAP-25) in vitro. Nesca expression in neurons is highly punctuate, co-stains with syntaxin-1, and is found in fractions containing markers of early endosomes and Golgi suggesting that it is involved in vesicular transport. Collectively, these data suggest that Nesca functions as an adapter involved in neuronal vesicular transport including vesicles containing soluble N-ethylmaleimide sensitive factor attachment protein receptors that are essential to exocytosis.
Collapse
Affiliation(s)
- James I S MacDonald
- Laboratory of Neural Signaling, Molecular Brain Research Group, Robarts Research Institute, Schulich School of Medicine, University of Western Ontario, London, ON, Canada
| | | | | | | | | | | |
Collapse
|
44
|
Nakamura N. The Role of the Transmembrane RING Finger Proteins in Cellular and Organelle Function. MEMBRANES 2011; 1:354-93. [PMID: 24957874 PMCID: PMC4021871 DOI: 10.3390/membranes1040354] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 11/24/2011] [Accepted: 12/05/2011] [Indexed: 01/08/2023]
Abstract
A large number of RING finger (RNF) proteins are present in eukaryotic cells and the majority of them are believed to act as E3 ubiquitin ligases. In humans, 49 RNF proteins are predicted to contain transmembrane domains, several of which are specifically localized to membrane compartments in the secretory and endocytic pathways, as well as to mitochondria and peroxisomes. They are thought to be molecular regulators of the organization and integrity of the functions and dynamic architecture of cellular membrane and membranous organelles. Emerging evidence has suggested that transmembrane RNF proteins control the stability, trafficking and activity of proteins that are involved in many aspects of cellular and physiological processes. This review summarizes the current knowledge of mammalian transmembrane RNF proteins, focusing on their roles and significance.
Collapse
Affiliation(s)
- Nobuhiro Nakamura
- Department of Biological Sciences, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan.
| |
Collapse
|
45
|
Iyengar PV, Hirota T, Hirose S, Nakamura N. Membrane-associated RING-CH 10 (MARCH10 protein) is a microtubule-associated E3 ubiquitin ligase of the spermatid flagella. J Biol Chem 2011; 286:39082-90. [PMID: 21937444 DOI: 10.1074/jbc.m111.256875] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Spermiogenesis is a complex and dynamic process of the metamorphosis of spermatids into spermatozoa. There is a great deal that is still unknown regarding the regulatory mechanisms for the formation of the sperm flagellum. In this study, we determined that the membrane-associated RING-CH 10 (March10) gene is predominantly expressed in rat testis. We isolated two March10 isoforms encoding MARCH10a and MARCH10b, which are generated by alternative splicing. MARCH10a is a long RING finger protein, and MARCH10b is a short RING finger-less protein. Immunohistochemical staining revealed that the MARCH10 proteins are specifically expressed in elongating and elongated spermatids, and the expression is absent in epididymal spermatozoa. MARCH10 immunoreactivity was observed in the cytoplasmic lobes as well as the principal piece and annulus of the flagella. When overexpressed in COS7 cells, MARCH10a was localized along the microtubules, whereas MARCH10b was distributed throughout the cytoplasm. An in vitro microtubule cosedimentation assay showed that MARCH10a is directly associated with microtubules. An in vitro ubiquitination assay demonstrated that the RING finger domain of MARCH10a exhibits an E3 ubiquitin ligase activity along with the E2 ubiquitin-conjugating enzyme UBE2B. Moreover, MARCH10a undergoes proteasomal degradation by autoubiquitination in transfected COS7 cells, but this activity was abolished upon microtubule disassembly. These results suggest that MARCH10 is involved in spermiogenesis by regulating the formation and maintenance of the flagella in developing spermatids.
Collapse
Affiliation(s)
- Prasanna Vasudevan Iyengar
- Department of Biological Sciences, Tokyo Institute of Technology, 4259-B-19 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | | | | | | |
Collapse
|
46
|
Eyster CA, Cole NB, Petersen S, Viswanathan K, Früh K, Donaldson JG. MARCH ubiquitin ligases alter the itinerary of clathrin-independent cargo from recycling to degradation. Mol Biol Cell 2011; 22:3218-30. [PMID: 21757542 PMCID: PMC3164467 DOI: 10.1091/mbc.e10-11-0874] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The MARCH family of proteins are membrane-associated E3 ubiquitin ligases that down-regulate surface membrane proteins. Expression of MARCH8 in cells causes the ubiquitination and down-regulation of surface CD98 and CD44—cargo proteins that enter cells by clathrin-independent endocytosis and are usually routed to recycling, not degradation. Following endocytosis, internalized plasma membrane proteins can be recycled back to the cell surface or trafficked to late endosomes/lysosomes for degradation. Here we report on the trafficking of multiple proteins that enter cells by clathrin-independent endocytosis (CIE) and determine that a set of proteins (CD44, CD98, and CD147) found primarily in recycling tubules largely failed to reach late endosomes in HeLa cells, whereas other CIE cargo proteins, including major histocompatibility complex class I protein (MHCI), trafficked to both early endosome antigen 1 (EEA1) and late endosomal compartments in addition to recycling tubules. Expression of the membrane-associated RING-CH 8 (MARCH8) E3 ubiquitin ligase completely shifted the trafficking of CD44 and CD98 proteins away from recycling tubules to EEA1 compartments and late endosomes, resulting in reduced surface levels. Cargo affected by MARCH expression, including CD44, CD98, and MHCI, still entered cells by CIE, suggesting that the routing of ubiquitinated cargo occurs after endocytosis. MARCH8 expression led to direct ubiquitination of CD98 and routing of CD98 to late endosomes/lysosomes.
Collapse
Affiliation(s)
- Craig A Eyster
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
47
|
Boname JM, Lehner PJ. What has the study of the K3 and K5 viral ubiquitin E3 ligases taught us about ubiquitin-mediated receptor regulation? Viruses 2011; 3:118-131. [PMID: 22049306 PMCID: PMC3206601 DOI: 10.3390/v3020118] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 01/17/2011] [Accepted: 01/20/2011] [Indexed: 12/29/2022] Open
Abstract
Cells communicate with each other and the outside world through surface receptors, which need to be tightly regulated to prevent both overstimulation and receptor desensitization. Understanding the processes involved in the homeostatic control of cell surface receptors is essential, but we are not alone in trying to regulate these receptors. Viruses, as the ultimate host pathogens, have co-evolved over millions of years and have both pirated and adapted host genes to enable viral pathogenesis. K3 and K5 (also known as MIR1 and MIR2) are viral ubiquitin E3 ligases from Kaposi's Sarcoma Associated Herpesvirus (KSHV) which decrease expression of a number of cell surface receptors and have been used to interrogate cellular processes and improve our understanding of ubiquitin-mediated receptor endocytosis and degradation. In this review, we summarize what has been learned from the study of these viral genes and emphasize their role in elucidating the complexity of ubiquitin in receptor regulation.
Collapse
Affiliation(s)
- Jessica M. Boname
- School of Clinical Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK; E-Mail:
| | - Paul J. Lehner
- School of Clinical Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK; E-Mail:
| |
Collapse
|
48
|
Bartee E, Eyster CA, Viswanathan K, Mansouri M, Donaldson JG, Früh K. Membrane-Associated RING-CH proteins associate with Bap31 and target CD81 and CD44 to lysosomes. PLoS One 2010; 5:e15132. [PMID: 21151997 PMCID: PMC2996310 DOI: 10.1371/journal.pone.0015132] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Accepted: 10/25/2010] [Indexed: 02/03/2023] Open
Abstract
Membrane-associated RING-CH (MARCH) proteins represent a family of transmembrane ubiquitin ligases modulating intracellular trafficking and turnover of transmembrane protein targets. While homologous proteins encoded by gamma-2 herpesviruses and leporipoxviruses have been studied extensively, limited information is available regarding the physiological targets of cellular MARCH proteins. To identify host cell proteins targeted by the human MARCH-VIII ubiquitin ligase we used stable isotope labeling of amino-acids in cell culture (SILAC) to monitor MARCH-dependent changes in the membrane proteomes of human fibroblasts. Unexpectedly, we observed that MARCH-VIII reduced the surface expression of Bap31, a chaperone that predominantly resides in the endoplasmic reticulum (ER). We demonstrate that Bap31 associates with the transmembrane domains of several MARCH proteins and controls intracellular transport of MARCH proteins. In addition, we observed that MARCH-VIII reduced the surface expression of the hyaluronic acid-receptor CD44 and both MARCH-VIII and MARCH-IV sequestered the tetraspanin CD81 in endo-lysosomal vesicles. Moreover, gene knockdown of MARCH-IV increased surface levels of endogenous CD81 suggesting a constitutive involvement of this family of ubiquitin ligases in the turnover of tetraspanins. Our data thus suggest a role of MARCH-VIII and MARCH-IV in the regulated turnover of CD81 and CD44, two ubiquitously expressed, multifunctional proteins.
Collapse
Affiliation(s)
- Eric Bartee
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Craig A. Eyster
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Kasinath Viswanathan
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Mandana Mansouri
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Julie G. Donaldson
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- * E-mail:
| |
Collapse
|
49
|
Nice TJ, Deng W, Coscoy L, Raulet DH. Stress-regulated targeting of the NKG2D ligand Mult1 by a membrane-associated RING-CH family E3 ligase. THE JOURNAL OF IMMUNOLOGY 2010; 185:5369-76. [PMID: 20870941 DOI: 10.4049/jimmunol.1000247] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
NKG2D is a stimulatory receptor expressed by NK cells and some T cell subsets. Expression of the self-encoded ligands for NKG2D is presumably tightly regulated to prevent autoimmune disorders while allowing detection of infected cells and developing tumors. The NKG2D ligand Mult1 is regulated at multiple levels, with a final layer of regulation controlling protein stability. In this article, we report that Mult1 cell-surface expression was prevented by two closely related E3 ubiquitin ligases membrane-associated RING-CH (MARCH)4 and MARCH9, members of an E3 family that regulates other immunologically active proteins. Lysines within the cytoplasmic domain of Mult1 were essential for this repression by MARCH4 or MARCH9. Downregulation of Mult1 by MARCH9 was reversed by heat-shock treatment, which resulted in the dissociation of the two proteins and increased the amount of Mult1 at the cell surface. These results identify Mult1 as a target for the MARCH family of E3 ligases and show that induction of Mult1 in response to heat shock is due to regulated association with its E3 ligases.
Collapse
Affiliation(s)
- Timothy J Nice
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720-3200, USA
| | | | | | | |
Collapse
|
50
|
Saito K, Nakamura N, Ito Y, Hoshijima K, Esaki M, Zhao B, Hirose S. Identification of zebrafish Fxyd11a protein that is highly expressed in ion-transporting epithelium of the gill and skin and its possible role in ion homeostasis. Front Physiol 2010; 1:129. [PMID: 21423371 PMCID: PMC3059942 DOI: 10.3389/fphys.2010.00129] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Accepted: 08/07/2010] [Indexed: 11/30/2022] Open
Abstract
FXYD proteins, small single-transmembrane proteins, have been proposed to be auxiliary regulatory subunits of Na+–K+-ATPase and have recently been implied in ion osmoregulation of teleost fish. In freshwater (FW) fish, numerous ions are actively taken up through mitochondrion-rich cells (MRCs) of the gill and skin epithelia, using the Na+ electrochemical gradient generated by Na+–K+-ATPase. In the present study, to understand the molecular mechanism for the regulation of Na+–K+-ATPase in MRCs of FW fish, we sought to identify FXYD proteins expressed in MRCs of zebrafish. Reverse-transcriptase PCR studies of adult zebrafish tissues revealed that, out of eight fxyd genes found in zebrafish database, only zebrafish fxyd11 (zfxyd11) mRNA exhibited a gill-specific expression. Double immunofluorescence staining showed that zFxyd11 is abundantly expressed in MRCs rich in Na+–K+-ATPase (NaK-MRCs) but not in those rich in vacuolar-type H+-transporting ATPase. An in situ proximity ligation assay demonstrated its close association with Na+–K+-ATPase in NaK-MRCs. The zfxyd11 mRNA expression was detectable at 1 day postfertilization, and its expression levels in the whole larvae and adult gills were regulated in response to changes in environmental ionic concentrations. Furthermore, knockdown of zFxyd11 resulted in a significant increase in the number of Na+–K+-ATPase–positive cells in the larval skin. These results suggest that zFxyd11 may regulate the transport ability of NaK-MRCs by modulating Na+–K+-ATPase activity, and may be involved in the regulation of body fluid and electrolyte homeostasis.
Collapse
Affiliation(s)
- Kaori Saito
- Department of Biological Sciences, Tokyo Institute of Technology Yokohama, Japan
| | | | | | | | | | | | | |
Collapse
|