1
|
Zheng Q, Li M, Qiu Y, Yang J, Cao Y. Overexpression of SLC35F2 is a potential prognostic biomarker for lung adenocarcinoma. Heliyon 2024; 10:e23828. [PMID: 38187235 PMCID: PMC10767229 DOI: 10.1016/j.heliyon.2023.e23828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 12/05/2023] [Accepted: 12/13/2023] [Indexed: 01/09/2024] Open
Abstract
Objective To explore the potential clinical and prognostic significance of Homo sapiens solute carrier family 35 member F2 (SLC35F2) in the context of lung adenocarcinoma (LUAD). Methods The expression pattern of SLC35F2 in LUAD tissues and normal tissues was analyzed in The Cancer Genome Atlas (TCGA) datasets and validated in 12 pairs of fresh clinical LUAD tissues and their corresponding adjacent normal tissues using quantitative real-time PCR (qRT-PCR) and western blotting. Immunohistochemistry (IHC) was used to assess the protein expression of SLC35F2 in 60 paraffin-embedded LUAD tissues, and its associations with clinicopathological parameters were further examined. The prognostic significance of SLC35F2 mRNA expression was also evaluated using the Kaplan-Meier method, and Cox regression models in LUAD patients from the TCGA database. The potential utility of SLC35F2 as an indicator of recurrence or metastasis was explored through the follow-up of selected clinical LUAD cases. Lastly, gene set enrichment analysis (GSEA) was conducted to investigate the underlying biological mechanisms and signaling pathways. Results Bioinformatics analysis utilizing the TCGA database indicated that SLC35F2 mRNA exhibited heightened expression in LUAD tissues when compared to normal tissues. These findings were further substantiated through the examination of 12 pairs of clinical LUAD tissues and their corresponding adjacent normal tissues, employing qRT-PCR and western blotting techniques. IHC results from a cohort of 60 LUAD patients demonstrated an up-regulation of SLC35F2 in 38 out of 60 individuals (63.3 %), which exhibited a significant correlation with tumor size, lymph node metastasis, and clinical stage (all P < 0.05). Both the Kaplan-Meier curve and the Cox proportional hazard analyses indicated a strong association between the up-regulation of SLC35F2 mRNA expression and unfavorable overall survival (OS) in patients with LUAD, as observed in the TCGA datasets (P < 0.05). The follow-up findings from select clinical LUAD cases provided evidence that the expression of SLC35F2 could serve as a dependable biomarker for monitoring the recurrence or metastasis. Additionally, the GSEA highlighted the enrichment of apoptosis, adhesion, small cell lung cancer (SCLC), and p53 signaling pathways in the subgroup of LUAD patients with elevated SLC35F2 expression. Conclusion SLC35F2 exhibited an up-regulated in both mRNA and protein expression, rendering it a valuable independent prognostic indicator for patients diagnosed with LUAD.
Collapse
Affiliation(s)
- Qingzhu Zheng
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Mingjie Li
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Yingkun Qiu
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Jiahao Yang
- Department of Laboratory Medicine, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, 350004, China
| | - Yingping Cao
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| |
Collapse
|
2
|
Wang Z, Yang D, Jiang Y, Wang Y, Niu M, Wang C, Luo H, Xu H, Li J, Zhang YW, Zhang X. Loss of RAB39B does not alter MPTP-induced Parkinson's disease-like phenotypes in mice. Front Aging Neurosci 2023; 15:1087823. [PMID: 36761179 PMCID: PMC9905435 DOI: 10.3389/fnagi.2023.1087823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/05/2023] [Indexed: 01/26/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative movement disorder with undetermined etiology. A major pathological hallmark of PD is the progressive degeneration of dopaminergic neurons in the substantia nigra. Loss-of-function mutations in the RAB39B gene, which encodes a neuronal-specific small GTPase RAB39B, have been associated with X-linked intellectual disability and pathologically confirmed early-onset PD in multiple families. However, the role of RAB39B in PD pathogenesis remains elusive. In this study, we treated Rab39b knock-out (KO) mice with MPTP to explore whether RAB39B deficiency could alter MPTP-induced behavioral impairments and dopaminergic neuron degeneration. Surprisingly, we found that MPTP treatment impaired motor activity and led to loss of tyrosine hydroxylase-positive dopaminergic neurons and gliosis in both WT and Rab39b KO mice. However, RAB39B deficiency did not alter MPTP-induced impairments. These results suggest that RAB39B deficiency does not contribute to PD-like phenotypes through compromising dopaminergic neurons in mice; and its role in PD requires further scrutiny.
Collapse
Affiliation(s)
- Zijie Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Xiamen University, Xiamen, China,Department of Neurosurgery, Xiang’an Hospital of Xiamen University, Xiamen, China
| | - Dingting Yang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Xiamen University, Xiamen, China,Department of Neurosurgery, Xiang’an Hospital of Xiamen University, Xiamen, China
| | - Yiru Jiang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Xiamen University, Xiamen, China
| | - Yong Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Xiamen University, Xiamen, China
| | - Mengxi Niu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Xiamen University, Xiamen, China
| | - Chong Wang
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
| | - Hong Luo
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Xiamen University, Xiamen, China
| | - Huaxi Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Xiamen University, Xiamen, China
| | - Jingwen Li
- Department of Neurosurgery, Xiang’an Hospital of Xiamen University, Xiamen, China
| | - Yun-wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Xiamen University, Xiamen, China
| | - Xian Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Center for Brain Sciences, The First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Xiamen University, Xiamen, China,*Correspondence: Xian Zhang, ✉
| |
Collapse
|
3
|
Kotolloshi R, Hölzer M, Gajda M, Grimm MO, Steinbach D. SLC35F2, a Transporter Sporadically Mutated in the Untranslated Region, Promotes Growth, Migration, and Invasion of Bladder Cancer Cells. Cells 2021; 10:E80. [PMID: 33418944 PMCID: PMC7825079 DOI: 10.3390/cells10010080] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/29/2020] [Accepted: 12/30/2020] [Indexed: 12/22/2022] Open
Abstract
Bladder cancer is a very heterogeneous disease and the molecular mechanisms of carcinogenesis and progression are insufficiently investigated. From the DNA sequencing analysis of matched non-muscle-invasive bladder cancer (NMIBC) and muscle-invasive bladder cancer (MIBC) samples from eight patients, we identified the tumour-associated gene SLC35F2 to be mutated in the 5' and 3' untranslated region (UTR). One mutation in 3'UTR increased the luciferase activity reporter, suggesting its influence on the protein expression of SLC35F2. The mRNA level of SLC35F2 was increased in MIBC compared with NMIBC. Furthermore, in immunohistochemical staining, we observed a strong intensity of SLC35F2 in single tumour cells and in the border cells of solid tumour areas with an atypical accumulation around the nucleus, especially in the MIBC. This suggests that SLC35F2 might be highly expressed in aggressive and invasive tumour cells. Moreover, knockdown of SLC35F2 repressed the growth of bladder cancer cells in the monolayer and spheroid model and suppressed migration and invasion of bladder cancer cells. In conclusion, we suggest that SLC35F2 is involved in bladder cancer progression and might provide a new therapeutic approach, for example, by the anti-cancer drug YM155, a cargo of the SLC35F2 transporter.
Collapse
Affiliation(s)
- Roland Kotolloshi
- Department of Urology, Jena University Hospital, 07740 Jena, Germany; (R.K.); (M.-O.G.)
| | - Martin Hölzer
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University Jena, 07743 Jena, Germany;
| | - Mieczyslaw Gajda
- Department of Forensic Medicine, Section of Pathology, Jena University Hospital, 07740 Jena, Germany;
| | - Marc-Oliver Grimm
- Department of Urology, Jena University Hospital, 07740 Jena, Germany; (R.K.); (M.-O.G.)
| | - Daniel Steinbach
- Department of Urology, Jena University Hospital, 07740 Jena, Germany; (R.K.); (M.-O.G.)
| |
Collapse
|
4
|
Tang BL. RAB39B's role in membrane traffic, autophagy, and associated neuropathology. J Cell Physiol 2020; 236:1579-1592. [PMID: 32761840 DOI: 10.1002/jcp.29962] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/19/2020] [Accepted: 07/13/2020] [Indexed: 12/14/2022]
Abstract
Neuropathological disorders are increasingly associated with dysfunctions in neuronal membrane traffic and autophagy, with defects among members of the Rab family of small GTPases implicated. Mutations in the human Xq28 localized gene RAB39B have been associated with X-linked neurodevelopmental defects including macrocephaly, intellectual disability, autism spectrum disorder (ASD), as well as rare cases of early-onset Parkinson's disease (PD). Despite the finding that RAB39B regulates GluA2 trafficking and could thus influence synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subunit composition, reasons for the wide-ranging neuropathological consequences associated with RAB39B defects have been unclear. Recent studies have now unraveled possible mechanisms underlying the neuropathological roles of this brain-enriched small GTPase. Studies in RAB39B knockout mice showed that RAB39B interacts with components of Class I phosphatidylinositol-3-kinase (PI3K) signaling. In its absence, the PI3K-AKT-mechanistic target of rapamycin signaling pathway in neural progenitor cells (NPCs) is hyperactivated, which promotes NPC proliferation, leading to macrocephaly and ASD. Pertaining to early-onset PD, a complex of C9orf72, Smith-Magenis syndrome chromosome region candidate 8 and WD repeat domain 41 that functions in autophagy has been identified as a guanine nucleotide exchange factor of RAB39B. Here, recent findings that have shed light on our mechanistic understanding of RAB39B's role in neurodevelopmental and neurodegenerative pathologies are reviewed. Caveats and unanswered questions are also discussed, and future perspectives outlined.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,NUS Graduate School of Integrative Sciences and Engineering, National University of Singapore, Singapore
| |
Collapse
|
5
|
Chen M, Gao X, Huang D, Wang S, Zheng L, Chen Y, Wen X, Gao Y, Cao H, Zhang S. Knockdown of SLC35F2 Inhibits the Proliferation and Metastasis of Bladder Cancer Cells. Onco Targets Ther 2019; 12:10771-10786. [PMID: 31849485 PMCID: PMC6911351 DOI: 10.2147/ott.s229332] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/19/2019] [Indexed: 01/03/2023] Open
Abstract
Background Many studies have shown that solute carrier family 35 member F2 (SLC35F2) plays a key role in the biological processes of multiple cancers. However, there have been no reports on the role of SLC35F2 in the occurrence and development of bladder cancer (BC). Methods SLC35F2 expression data and clinical and prognostic information from BC patients were obtained from databases. SLC35F2 expression in BC was verified by quantitative real-time PCR (qRT-PCR). The influence of SLC35F2 knockdown on the proliferation, apoptosis, migration and invasion in the 5637 and T24 cell lines was studied, and tumor formation experiments were performed in nude mice. Gene set enrichment analysis (GSEA) was used to predict the pathways and functions of SLC35F2 in BC. Results SLC35F2 was highly expressed in BC tissues and was associated with invasiveness and T stage in BC patients. SLC35F2 knockdown can inhibit the proliferation, migration and invasion of BC cells and can promote apoptosis. SLC35F2 knockdown significantly reduced tumorigenesis in nude mice. GSEA showed that BC, pathways in cancer, apoptosis and the P53 signaling pathway were significantly enriched in SLC35F2 high expression phenotype. Conclusion SLC35F2 can promote malignant progression and is a potential therapeutic target in BC.
Collapse
Affiliation(s)
- Mei Chen
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, People's Republic of China
| | - Xin Gao
- Clinical Laboratory, The First People's Hospital of Huaihua, Huaihua 418000, People's Republic of China
| | - Denggao Huang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, People's Republic of China
| | - Shunlan Wang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, People's Republic of China
| | - Linlin Zheng
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, People's Republic of China
| | - Yinyi Chen
- Clinical Laboratory, The First Affiliated Hospital of University of South China, Hengyang 421001, People's Republic of China
| | - Xiaohong Wen
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, People's Republic of China
| | - Yuanhui Gao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, People's Republic of China
| | - Hui Cao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, People's Republic of China
| | - Shufang Zhang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou 570208, People's Republic of China
| |
Collapse
|
6
|
He J, Jin Y, Zhou M, Li X, Chen W, Wang Y, Gu S, Cao Y, Chu C, Liu X, Zou Q. Solute carrier family 35 member F2 is indispensable for papillary thyroid carcinoma progression through activation of transforming growth factor-β type I receptor/apoptosis signal-regulating kinase 1/mitogen-activated protein kinase signaling axis. Cancer Sci 2018; 109:642-655. [PMID: 29274137 PMCID: PMC5834798 DOI: 10.1111/cas.13478] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/14/2017] [Accepted: 12/19/2017] [Indexed: 02/05/2023] Open
Abstract
Solute carrier family members control essential physiological functions and are tightly linked to human diseases. Solute carrier family 35 member F2 (SLC35F2) is aberrantly activated in several malignancies. However, the biological function and molecular mechanism of SLC35F2 in papillary thyroid carcinoma (PTC) are yet to be fully explored. Here, we showed that SLC35F2 was prominently upregulated in PTC tissues at both protein and mRNA expression level compared with matched adjacent normal tissues. Besides, the high expression of SLC35F2 was significantly associated with lymph node metastasis in patients with PTC. CRISPR/Cas9-mediated knockout of SLC35F2 attenuated the tumorigenic properties of PTC, including cell proliferation, migration and invasion and induced G1 phase arrest. In contrast, ectopic expression of SLC35F2 brought about aggressive malignant phenotypes of PTC cells. Moreover, SLC35F2 expedited the proliferation and migration of PTC cells by targeting transforming growth factor-β type I receptor (TGFBR1) and phosphorylation of apoptosis signal-regulating kinase 1 (p-ASK-1), thereby activating the mitogen-activated protein kinase signaling pathway. The malignant behaviors induced by overexpression of SLC35F2 could be abrogated by silencing of TGFBR1 using a specific inhibitor. We conducted the first study on SLC35F2 in thyroid cancer with the aim of elucidating the functional significance and molecular mechanism of SLC35F2. Our findings suggest that SLC35F2 exerts its oncogenic effect on PTC progression through the mitogen-activated protein kinase pathway, with dependence on activation of TGFBR-1 and apoptosis signal-regulating kinase 1.
Collapse
Affiliation(s)
- Jing He
- Department of General SurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Yiting Jin
- Department of General SurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Mingxia Zhou
- Department of GastroenterologyXinhua HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xiaoyan Li
- Department of General SurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Wanna Chen
- Department of General SurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Yiwei Wang
- Department of General SurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Siwen Gu
- Department of General SurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Yun Cao
- Department of General SurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Chengyu Chu
- Department of General SurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Xiuping Liu
- Department of PathologySchool of Basic Medical SciencesFudan UniversityShanghaiChina
- Department of PathologyThe Fifth People's Hospital of ShanghaiFudan UniversityShanghaiChina
| | - Qiang Zou
- Department of General SurgeryHuashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
7
|
Influence on the behavior of lung cancer H1299 cells by silencing SLC35F2 expression. Cancer Cell Int 2013; 13:73. [PMID: 23879892 PMCID: PMC3724707 DOI: 10.1186/1475-2867-13-73] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 07/15/2013] [Indexed: 11/17/2022] Open
Abstract
Background To investigate the effects of RNA interference-mediated downregulation of Human Solute Carrier Family 35 member F2 (SLC35F2) expression on the biological behavior of lung cancer H1299 cells. Methods The lentiviral vector of small interfering RNA targeting SLC35F2 was introduced into H1299 cells by liposome-mediated transfection. Expression of the SLC35F2 protein was measured by western blot. The proliferation of H1299 cells was determined by Cell Counting Kit-8 assay. The migration of H1299 cells was measured by Transwell migration assay. Cell cycle analysis used fluorescence-activated cell sorting. Results SLC35F2 expression was markedly downregulated in H1299 cell clone (transfected with the lentiviral vector harboring small interfering RNA targeting SLC35F2). Proliferation decreased significantly compared with that of non-transfected H1299 cells. Transwell migration assay showed that fewer cells moved through the artificial basement membrane compared with untransfected H1299 cells (38.3 ± 5.7 vs. 113.5 ± 8.5, P < 0.05). The cell cycle of H1299 cells was changed, the percentage of H1299 cells in S and G2/M phases being significantly decreased compared with untransfected H1299 cells (S phase: 15.3% ± 3.0% vs. 27.0% ± 5.4%, P > 0.05; G2/M phase; 3.0% ± 1.1% vs. 10.5% ± 1.7%, P < 0.05), whereas the percentage of H1299 cells in G0/G1 phase increased markedly (81.7% ± 4.0% vs. 62.5% ± 1.9%, P < 0.05). Conclusion RNA interference-mediated downregulation of SLC35F2 expression by lentiviral vector can attenuate the proliferation, migration and invasion of H1299 cells.
Collapse
|
8
|
Small GTPase Rab39A interacts with UACA and regulates the retinoic acid-induced neurite morphology of Neuro2A cells. Biochem Biophys Res Commun 2013; 435:113-9. [DOI: 10.1016/j.bbrc.2013.04.051] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 04/11/2013] [Indexed: 01/05/2023]
|
9
|
Abstract
The DENN domain is a common, evolutionarily ancient, and conserved protein module, yet it has gone largely unstudied; until recently, little was known regarding its functional roles. New studies reveal that various DENN domains interact directly with members of the Rab family of small GTPases and that DENN domains function enzymatically as Rab-specific guanine nucleotide exchange factors. Thus, DENN domain proteins appear to be generalized regulators of Rab function. Study of these proteins will provide new insights into Rab-mediated membrane trafficking pathways.
Collapse
Affiliation(s)
- Andrea L. Marat
- From the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Hatem Dokainish
- From the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Peter S. McPherson
- From the Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| |
Collapse
|
10
|
Becker CE, Creagh EM, O'Neill LAJ. Rab39a binds caspase-1 and is required for caspase-1-dependent interleukin-1beta secretion. J Biol Chem 2009; 284:34531-7. [PMID: 19833722 PMCID: PMC2787314 DOI: 10.1074/jbc.m109.046102] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Interleukin-1β (IL-1β) is an important pro-inflammatory cytokine that is secreted by unconventional means in a caspase-1-dependent manner. Using a one-step immunoprecipitation approach to isolate endogenous caspase-1 from the monocytic THP1 cell line, we identified previously undescribed binding partners using mass spectrometry. One of the proteins identified was Rab39a, a member of the Rab GTPase family, a group of proteins that have important roles in protein trafficking and secretion. We confirmed by co-immunoprecipitation that Rab39a binds caspase-1. Knock down of Rab39a with small interfering RNA resulted in diminished levels of secreted IL-1β but had no effect on induction of pro-IL-1β mRNA by lipopolysaccharide. Rab39a contains a highly conserved caspase-1 cleavage site and was cleaved in the presence of recombinant caspase-1 or lipopolysaccharide. Finally, overexpression of Rab39a results in an increase in IL-1β secretion, and furthermore, overexpression of a Rab39a construct lacking the caspase-1 cleavage site leads to an additional increase in IL-1β secretion. Altogether, our findings show that Rab39a interacts with caspase-1 and suggest that Rab39a functions as a trafficking adaptor linking caspase-1 to IL-1β secretion.
Collapse
Affiliation(s)
- Christine E Becker
- School of Biochemistry and Immunology, Trinity College, Dublin 2, Ireland
| | | | | |
Collapse
|
11
|
Lespinet V, Terraz F, Recher C, Campo E, Hall J, Delsol G, Al Saati T. Single-cell analysis of loss of heterozygosity at theATM gene locus in Hodgkin and Reed-Sternberg cells of Hodgkin's lymphoma:ATM loss of heterozygosity is a rare event. Int J Cancer 2005; 114:909-16. [PMID: 15645496 DOI: 10.1002/ijc.20825] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Hodgkin's lymphoma (HL) is a lymphoid malignancy characterized by the presence of rare neoplastic cells, Hodgkin and Reed-Sternberg (HRS) cells, scattered among a predominant population of inflammatory cells. On the basis of previously reported cytogenetic analyses, the ATM (ataxia-telangiectasia mutated) gene at 11q22-23 has been implicated in the etiology of HL. We therefore developed a single-cell PCR approach to detect ATM loss of heterozygosity (LOH) in HRS cells. Three microsatellites were investigated; 1 localized inside the ATM gene and the remaining 2 in close proximity. In 2 of the 15 lymph node samples, an allelic loss of the ATM gene locus was detected. ATM protein expression was examined in 8 cases (including 1 of the 2 cases with LOH) by immunohistochemistry. In the case associated with an allelic loss, the ATM protein was absent in the HRS cells, whereas in the 7 remaining cases, without detectable LOH at the ATM locus, nuclear ATM expression was observed. In the 2 HL cases with LOH, the ATM gene was sequenced following whole genome amplification of DNA isolated from microdissected HRS cells. In 1 of these 2 cases, a splice site mutation in the second ATM allele was found. This mutation could generate a premature termination codon leading to a marked instability and a rapid degradation of the resulting ATM mRNA transcripts. This latter event could explain the loss of the expression of the ATM protein in HRS cells as detected by immunohistochemistry in this particular case. As previously reported in some B-cell lymphomas, our results suggest that ATM genetic anomalies could play a role in the pathogenesis of a subset of HL cases.
Collapse
Affiliation(s)
- Virginie Lespinet
- INSERM U563, CPTP, Oncogenèse et Signalisation dans les Cellules Hématopoïétiques et Plateau Technique d'Histopathologie Experimentale, IFR30, Purpan Hospital, 31059 Toulouse, France
| | | | | | | | | | | | | |
Collapse
|
12
|
Shen C, Wang D, Jiang G, Wang J, Zhang G. Molecular cloning, identification and analysis of lung squamous cell carcinoma-related genes. Lung Cancer 2002; 38:235-41. [PMID: 12445744 DOI: 10.1016/s0169-5002(02)00300-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To clone and identify genes differentially expressed in human lung squamous cell carcinoma (LSCC). METHODS A subtracted cDNA library of human LSCC was constructed by using suppression subtractive hybridization (SSH) method. Through screening, the subtracted library clones, representing mRNAs that are truly differentially expressed in LSCC but not in normal lung tissues, were selected out to identify by semi-quantitative RT-PCR in 12 patients of LSCC and performed DNA sequencing. Nucleic acid homology searches were performed using the BLAST program. Partial novel genes were detected by Northern blot. RESULTS By this technique, we obtained 10 differentially expressed gene cDNA fragments of LSCC. Among them six were already known genes; two sequences were already identified but their functions were still unknown (hypothetical protein); two were novel (GenBank accession number were AF363068 and AY032661, respectively). The results from semi-quantitative RT-PCR showed that the transcription expression level of these clones including PPP1CB, caluminin, S100A2, HSNOV1, OCIA and AY032661 was down-regulated in 12 cases of LSCC, while the transcription of HSP90, ferritin, gp96 and AF363068 was up-regulated in same cases. CONCLUSION SSH is a powerful technique of high sensitivity for the detection of differential gene expression in LSCC and an effective method to clone novel genes. Six already known genes identified by SSH technique have been already implicated in the pathogenesis of lung carcinogenesis, or they are involved in immunological defense mechanism in human body. Two hypothetical proteins probably also play an important role in lung cancer pathogenesis. The function of two novel genes in lung carcinogenesis is under research.
Collapse
Affiliation(s)
- Chenyang Shen
- Department of Thoracic Surgery, People's Hospital, Peking University, Beijing 100044, China.
| | | | | | | | | |
Collapse
|
13
|
Pereira-Leal JB, Seabra MC. The mammalian Rab family of small GTPases: definition of family and subfamily sequence motifs suggests a mechanism for functional specificity in the Ras superfamily. J Mol Biol 2000; 301:1077-87. [PMID: 10966806 DOI: 10.1006/jmbi.2000.4010] [Citation(s) in RCA: 344] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The Rab/Ypt/Sec4 family forms the largest branch of the Ras superfamily of GTPases, acting as essential regulators of vesicular transport pathways. We used the large amount of information in the databases to analyse the mammalian Rab family. We defined Rab-conserved sequences that we designate Rab family (RabF) motifs using the conserved PM and G motifs as "landmarks". The Rab-specific regions were used to identify new Rab proteins in the databases and suggest rules for nomenclature. Surprisingly, we find that RabF regions cluster in and around switch I and switch II regions, i.e. the regions that change conformation upon GDP or GTP binding. This finding suggests that specificity of Rab-effector interaction cannot be conferred solely through the switch regions as is usually inferred. Instead, we propose a model whereby an effector binds to RabF (switch) regions to discriminate between nucleotide-bound states and simultaneously to other regions that confer specificity to the interaction, possibly Rab subfamily (RabSF) specific regions that we also define here. We discuss structural and functional data that support this model and its general applicability to the Ras superfamily of proteins.
Collapse
Affiliation(s)
- J B Pereira-Leal
- Molecular Genetics Section Division of Biomedical Sciences, Imperial College School of Medicine, Sir Alexander Fleming Building, London, SW7 2AZ, UK
| | | |
Collapse
|
14
|
Laake K, Launonen V, Niederacher D, Gudlaugsdottir S, Seitz S, Rio P, Champ�me MH, Bi�che I, Birnbaum D, White G, Sztan M, Sever N, Plummer S, Osorio A, Broeks A, Huusko P, Spurr N, Borg �, Cleton-Jansen AM, Veer LV, Benitez J, Casey G, Peterlin B, Olah E, Varley J, Bignon YJ, Scherneck S, Sigurdardottir V, Lidereau R, Eyfjord J, Beckmann MW, Winqvist R, Skovlund E, B�rresen-Dale AL. Loss of heterozygosity at 11q23.1 and survival in breast cancer: Results of a large European study. Genes Chromosomes Cancer 1999. [DOI: 10.1002/(sici)1098-2264(199907)25:3<212::aid-gcc3>3.0.co;2-g] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
15
|
Affiliation(s)
- J Slingerland
- Department of Pathology, New York University Medical Center, New York 10016, USA
| | | |
Collapse
|
16
|
Byrd PJ, Stankovic T, McConville CM, Smith AD, Cooper PR, Taylor AM. Identification and analysis of expression of human VACM-1, a cullin gene family member located on chromosome 11q22-23. Genome Res 1997; 7:71-5. [PMID: 9037604 DOI: 10.1101/gr.7.1.71] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
We have localized the human homolog of the rabbit vasopressin-activated calcium-mobilizing receptor VACM-1 to a region close to the gene for ataxia telangiectasia ATM on chromosome 11q22-23. We have determined the complete amino acid sequence of the human Hs-VACM-1 protein, which is 780 amino acids long. The human and rabbit sequences are highly conserved, differing at only seven amino acids. Northern analysis of the human gene showed expression in a wide range of human tissues. The Hs-VACM-1 gene has homology with the Caenorhabditis elegans gene Ce-cul-5, a member of a family of cullin genes that are involved in cell cycle regulation and that might, when mutated, contribute to tumor progression.
Collapse
|