1
|
Wang WJ, Lyu TJ, Li Z. Research Progress on PATJ and Underlying Mechanisms Associated with Functional Outcomes After Stroke. Neuropsychiatr Dis Treat 2021; 17:2811-2818. [PMID: 34471355 PMCID: PMC8405222 DOI: 10.2147/ndt.s310764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/24/2021] [Indexed: 12/05/2022] Open
Abstract
Cell polarity is an intrinsic property of epithelial cells regulated by scaffold proteins. The CRB (crumbs) complex is known to play a predominant role in the dynamic cooperative network of polarity scaffold proteins. PATJ (PALS1-associated tight junction) is the core component in the CRB complex and has been highly conserved throughout evolution. PATJ is crucial to several important events in organisms' survival, including embryonic development, cell polarity, and barrier establishment. A recent study shows that PATJ plays an important role in functional outcomes of stroke. In this article, we elaborate on the biological structure and physiological functions of PATJ and explore the underlying mechanisms of PATJ genetic polymorphism that are associated with poor functional outcomes in ischemic stroke.
Collapse
Affiliation(s)
- Wen-Jie Wang
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Tian-Jie Lyu
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, People's Republic of China.,National Center for Healthcare Quality Management in Neurological Diseases, Beijing, 100070, People's Republic of China
| | - Zixiao Li
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China.,China National Clinical Research Center for Neurological Diseases, Beijing, 100070, People's Republic of China.,National Center for Healthcare Quality Management in Neurological Diseases, Beijing, 100070, People's Republic of China.,Chinese Institute for Brain Research, Beijing, 100070, People's Republic of China.,Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, 100070, People's Republic of China
| |
Collapse
|
2
|
Hong J, Won M, Ro H. The Molecular and Pathophysiological Functions of Members of the LNX/PDZRN E3 Ubiquitin Ligase Family. Molecules 2020; 25:E5938. [PMID: 33333989 PMCID: PMC7765395 DOI: 10.3390/molecules25245938] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/10/2020] [Accepted: 12/10/2020] [Indexed: 12/27/2022] Open
Abstract
The ligand of Numb protein-X (LNX) family, also known as the PDZRN family, is composed of four discrete RING-type E3 ubiquitin ligases (LNX1, LNX2, LNX3, and LNX4), and LNX5 which may not act as an E3 ubiquitin ligase owing to the lack of the RING domain. As the name implies, LNX1 and LNX2 were initially studied for exerting E3 ubiquitin ligase activity on their substrate Numb protein, whose stability was negatively regulated by LNX1 and LNX2 via the ubiquitin-proteasome pathway. LNX proteins may have versatile molecular, cellular, and developmental functions, considering the fact that besides these proteins, none of the E3 ubiquitin ligases have multiple PDZ (PSD95, DLGA, ZO-1) domains, which are regarded as important protein-interacting modules. Thus far, various proteins have been isolated as LNX-interacting proteins. Evidence from studies performed over the last two decades have suggested that members of the LNX family play various pathophysiological roles primarily by modulating the function of substrate proteins involved in several different intracellular or intercellular signaling cascades. As the binding partners of RING-type E3s, a large number of substrates of LNX proteins undergo degradation through ubiquitin-proteasome system (UPS) dependent or lysosomal pathways, potentially altering key signaling pathways. In this review, we highlight recent and relevant findings on the molecular and cellular functions of the members of the LNX family and discuss the role of the erroneous regulation of these proteins in disease progression.
Collapse
Affiliation(s)
- Jeongkwan Hong
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 305-764, Korea;
| | - Minho Won
- Biotechnology Process Engineering Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 30 Yeongudanji-ro, Cheongwon-gu, Cheongju 28116, Korea
| | - Hyunju Ro
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 305-764, Korea;
| |
Collapse
|
3
|
Huang Y, Huang X, Cheng C, Xu X, Liu H, Yang X, Yao L, Ding Z, Tang J, He S, Wang Y. Elucidating the expression and function of Numbl during cell adhesion-mediated drug resistance (CAM-DR) in multiple myeloma (MM). BMC Cancer 2019; 19:1269. [PMID: 31888545 PMCID: PMC6937660 DOI: 10.1186/s12885-019-6446-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 12/09/2019] [Indexed: 12/17/2022] Open
Abstract
Background Cell adhesion-mediated drug resistance (CAM-DR) is a major clinical problem that prevents successful treatment of multiple myeloma (MM). In particular, the expression levels of integrin β1 and its sub-cellular distribution (internalization and trafficking) are strongly associated with CAM-DR development. Methods Development of an adhesion model of established MM cell lines and detection of Numbl and Integrinβ1 expression by Western Blot analysis. The interaction between Numbl and Integrinβ1 was assessed by a co-immunoprecipitation (CO-IP) method. Calcein AM assay was performed to investigate the levels of cell adhesion. Finally, the extent of CAM-DR in myeloma cells was measured using cell viability assay and flow cytometry analysis. Results Our preliminary date suggest that Numbl is differentially expressed in a cell adhesion model of MM cell lines. In addition to binding to the phosphotyrosine-binding (PTB) domain, the carboxyl terminal of Numbl can also interact with integrin β1 to regulate the cell cycle by activating the pro-survival PI3K/AKT signaling pathway. This study intends to verify and elucidate the interaction between Numbl and integrin β1 and its functional outcome on CAM-DR. We have designed and developed a CAM-DR model using MM cells coated with either fibronectin or bone marrow stromal cells. We assessed whether Numbl influences cell-cycle progression and whether it, in turn, contributes to activation of PI3K/AKT signal pathway through the adjustment of its carboxyl end. Finally, we showed that the interaction of Numbl with integrin β1 promotes the formation of CAM-DR in MM cells. Conclusions Our findings elucidated the specific molecular mechanisms of CAM-DR induction and confirmed that Numbl is crucial for the development of CAM-DR in MM cells.
Collapse
Affiliation(s)
- Yuejiao Huang
- Department of Oncology, Nantong University Cancer Hospital, Nantong, Jiangsu, 226001, People's Republic of China
| | - Xianting Huang
- Department of Oncology center, Jiangsu Jiangyin People's Hospital, Jiangyin, Jiangsu, 214400, People's Republic of China
| | - Chun Cheng
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Xiaohong Xu
- Department of Oncology, Nantong University Cancer Hospital, Nantong, Jiangsu, 226001, People's Republic of China
| | - Hong Liu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Xiaojing Yang
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Li Yao
- Department of Immunology, Medical College of Jiangnan University, Wuxi, Jiangsu, 214122, People's Republic of China
| | - Zongmei Ding
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Jie Tang
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Song He
- Department of Pathology, Nantong University Cancer Hospital, Nantong, Jiangsu, 226001, People's Republic of China.
| | - Yuchan Wang
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China.
| |
Collapse
|
4
|
LNX1/LNX2 proteins: functions in neuronal signalling and beyond. Neuronal Signal 2018; 2:NS20170191. [PMID: 32714586 PMCID: PMC7373230 DOI: 10.1042/ns20170191] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/02/2018] [Accepted: 05/08/2018] [Indexed: 12/16/2022] Open
Abstract
Ligand of NUMB Protein X1 and X2 (LNX1 and LNX2) are E3 ubiquitin ligases, named for their ability to interact with and promote the degradation of the cell fate determinant protein NUMB. On this basis they are thought to play a role in modulating NUMB/NOTCH signalling during processes such as cortical neurogenesis. However, LNX1/2 proteins can bind, via their four PDZ (PSD95, DLGA, ZO-1) domains, to an extraordinarily large number of other proteins besides NUMB. Many of these interactions suggest additional roles for LNX1/2 proteins in the nervous system in areas such as synapse formation, neurotransmission and regulating neuroglial function. Twenty years on from their initial discovery, I discuss here the putative neuronal functions of LNX1/2 proteins in light of the anxiety-related phenotype of double knockout mice lacking LNX1 and LNX2 in the central nervous system (CNS). I also review what is known about non-neuronal roles of LNX1/2 proteins, including their roles in embryonic patterning and pancreas development in zebrafish and their possible involvement in colorectal cancer (CRC), osteoclast differentiation and immune function in mammals. The emerging picture places LNX1/2 proteins as potential regulators of multiple cellular signalling processes, but in many cases the physiological significance of such roles remains only partly validated and needs to be considered in the context of the tight control of LNX1/2 protein levels in vivo.
Collapse
|
5
|
Liu TT, Achrol AS, Mitchell LA, Du WA, Loya JJ, Rodriguez SA, Feroze A, Westbroek EM, Yeom KW, Stuart JM, Chang SD, Harsh GR, Rubin DL. Computational Identification of Tumor Anatomic Location Associated with Survival in 2 Large Cohorts of Human Primary Glioblastomas. AJNR Am J Neuroradiol 2016; 37:621-8. [PMID: 26744442 DOI: 10.3174/ajnr.a4631] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/02/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE Tumor location has been shown to be a significant prognostic factor in patients with glioblastoma. The purpose of this study was to characterize glioblastoma lesions by identifying MR imaging voxel-based tumor location features that are associated with tumor molecular profiles, patient characteristics, and clinical outcomes. MATERIALS AND METHODS Preoperative T1 anatomic MR images of 384 patients with glioblastomas were obtained from 2 independent cohorts (n = 253 from the Stanford University Medical Center for training and n = 131 from The Cancer Genome Atlas for validation). An automated computational image-analysis pipeline was developed to determine the anatomic locations of tumor in each patient. Voxel-based differences in tumor location between good (overall survival of >17 months) and poor (overall survival of <11 months) survival groups identified in the training cohort were used to classify patients in The Cancer Genome Atlas cohort into 2 brain-location groups, for which clinical features, messenger RNA expression, and copy number changes were compared to elucidate the biologic basis of tumors located in different brain regions. RESULTS Tumors in the right occipitotemporal periventricular white matter were significantly associated with poor survival in both training and test cohorts (both, log-rank P < .05) and had larger tumor volume compared with tumors in other locations. Tumors in the right periatrial location were associated with hypoxia pathway enrichment and PDGFRA amplification, making them potential targets for subgroup-specific therapies. CONCLUSIONS Voxel-based location in glioblastoma is associated with patient outcome and may have a potential role for guiding personalized treatment.
Collapse
Affiliation(s)
- T T Liu
- From the Stanford Center for Biomedical Informatics Research and Biomedical Informatics Training Program (T.T.L., D.L.R.) Department of Radiology (T.T.L., L.A.M., W.A.D., K.W.Y., D.L.R.)
| | - A S Achrol
- Stanford Institute for Neuro-Innovation and Translational Neurosciences (A.S.A.) Institute for Stem Cell Biology and Regenerative Medicine (A.S.A.) Department of Neurosurgery (A.S.A., J.J.L., S.A.R., E.M.W., S.D.C., G.R.H.), Stanford University School of Medicine, Stanford, California
| | - L A Mitchell
- Department of Radiology (T.T.L., L.A.M., W.A.D., K.W.Y., D.L.R.)
| | - W A Du
- Department of Radiology (T.T.L., L.A.M., W.A.D., K.W.Y., D.L.R.)
| | - J J Loya
- Department of Neurosurgery (A.S.A., J.J.L., S.A.R., E.M.W., S.D.C., G.R.H.), Stanford University School of Medicine, Stanford, California
| | - S A Rodriguez
- Department of Neurosurgery (A.S.A., J.J.L., S.A.R., E.M.W., S.D.C., G.R.H.), Stanford University School of Medicine, Stanford, California
| | - A Feroze
- Department of Neurological Surgery (A.F.), University of Washington School of Medicine, Seattle, Washington
| | - E M Westbroek
- Department of Neurosurgery (A.S.A., J.J.L., S.A.R., E.M.W., S.D.C., G.R.H.), Stanford University School of Medicine, Stanford, California
| | - K W Yeom
- Department of Radiology (T.T.L., L.A.M., W.A.D., K.W.Y., D.L.R.)
| | - J M Stuart
- Biomolecular Engineering (J.M.S.), University of California Santa Cruz, Santa Cruz, California
| | - S D Chang
- Department of Neurosurgery (A.S.A., J.J.L., S.A.R., E.M.W., S.D.C., G.R.H.), Stanford University School of Medicine, Stanford, California
| | - G R Harsh
- Department of Neurosurgery (A.S.A., J.J.L., S.A.R., E.M.W., S.D.C., G.R.H.), Stanford University School of Medicine, Stanford, California
| | - D L Rubin
- From the Stanford Center for Biomedical Informatics Research and Biomedical Informatics Training Program (T.T.L., D.L.R.) Department of Radiology (T.T.L., L.A.M., W.A.D., K.W.Y., D.L.R.)
| |
Collapse
|
6
|
Hu T, Yang H, Han ZG. PDZRN4 acts as a suppressor of cell proliferation in human liver cancer cell lines. Cell Biochem Funct 2015; 33:443-9. [PMID: 26486104 DOI: 10.1002/cbf.3130] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/12/2015] [Accepted: 08/03/2015] [Indexed: 11/11/2022]
Abstract
Recently, some reports show that Ligand of Numb Protein-X 1 (LNX1) could be a suppressor gene in gliomas, while our current research has firstly shown that PDZ domain containing ring finger 4 (PDZRN4), another member of LNX family, could also be a potential suppressor in hepatocellular carcinoma (HCC). PDZRN4, also named LNX4 (Ligand of Numb Protein-X 4), is a member of the LNX family. We recently found that PDZRN4, but not LNX1, was down-regulated in HCC samples, and the role of PDZRN4 in the progression of HCC had not been studied before. To address this question, firstly, we evaluated the expression of PDZRN4 in HCC samples and adjacent non-cancerous tissues. Semi-quantitative polymerase chain reaction showed that PDZRN4 was down-regulated in 24/36 (66.7%) HCC samples separately. In addition, our research shows that PDZRN4 is silenced in all of the 12 HCC cell lines tested. Subsequently, cell-based functional assay exhibited that ectopic expression of PDZRN4 inhibits the proliferation, plate colony formation and anchorage-independent colony formation of HCC cells. Collectively, our results showed that PDZRN4 might be a potential tumour suppressor gene and had anti-proliferative effect on HCC cell proliferation, which would be of great significance to the researches on HCC.
Collapse
Affiliation(s)
- Taotao Hu
- Key Laboratory of Systems Biomedicine (Ministry of Education) of Rui-Jin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center, Shanghai, Shanghai, China
| | - Hong Yang
- Key Laboratory of Systems Biomedicine (Ministry of Education) of Rui-Jin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Ze-Guang Han
- Key Laboratory of Systems Biomedicine (Ministry of Education) of Rui-Jin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Center for Systems Biomedicine, Shanghai Jiaotong University, Shanghai, China.,Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center, Shanghai, Shanghai, China
| |
Collapse
|
7
|
Lenihan JA, Saha O, Mansfield LM, Young PW. Tight, cell type-specific control of LNX expression in the nervous system, at the level of transcription, translation and protein stability. Gene 2014; 552:39-50. [PMID: 25200495 DOI: 10.1016/j.gene.2014.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 09/01/2014] [Accepted: 09/04/2014] [Indexed: 11/19/2022]
Abstract
LNX1 and LNX2 are E3 ubiquitin ligases that can interact with Numb - a key regulator of neurogenesis and neuronal differentiation. LNX1 can target Numb for proteasomal degradation, and Lnx mRNAs are prominently expressed in the nervous system, suggesting that LNX proteins play a role in neural development. This hypothesis remains unproven, however, largely because LNX proteins are present at very low levels in vivo. Here, we demonstrate expression of both LNX1 and LNX2 proteins in the brain for the first time. We clarify the cell-type specific expression of LNX isoforms in both the CNS and PNS, and identify a novel LNX1 isoform. Using luciferase reporter assays, we show that the 5' untranslated region of the Lnx1_variant 2 mRNA, that generates the LNX1p70 isoform, strongly suppresses protein production. This effect is mediated in part by the presence of upstream open reading frames (uORFs), but also by a sequence element that decreases both mRNA levels and translational efficiency. By contrast, uORFs do not negatively regulate LNX1p80 or LNX2 expression. Instead, we find some evidence that protein turnover via proteasomal degradation may influence LNX1p80 levels in cells. These observations provide plausible explanations for the low levels of LNX1 proteins detected in vivo.
Collapse
Affiliation(s)
- Joan A Lenihan
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Orthis Saha
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Louise M Mansfield
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Paul W Young
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland.
| |
Collapse
|
8
|
Guo Z, Song E, Ma S, Wang X, Gao S, Shao C, Hu S, Jia L, Tian R, Xu T, Gao Y. Proteomics strategy to identify substrates of LNX, a PDZ domain-containing E3 ubiquitin ligase. J Proteome Res 2012; 11:4847-4862. [PMID: 22889411 DOI: 10.1021/pr300674c] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ubiquitin ligases (E3s) confer specificity to ubiquitination by recognizing target substrates. However, the substrates of most E3s have not been extensively discovered, and new methods are needed to efficiently and comprehensively identify these substrates. Mostly, E3s specifically recognize substrates via their protein interaction domains. We developed a novel integrated strategy to identify substrates of E3s containing protein interaction domains on a proteomic scale. The binding properties of the protein interaction domains were characterized by screening a random peptide library using a yeast two-hybrid system. Artificial degrons, consisting of a preferential ubiquitination sequence and particular interaction domain-binding motifs, were tested as potential substrates by in vitro ubiquitination assays. Using this strategy, not only substrates but also nonsubstrate regulators can be discovered. The detailed substrate recognition mechanisms, which are useful for drug discovery, can also be characterized. We used the Ligand of Numb protein X (LNX) family of E3s, a group of PDZ domain-containing RING-type E3 ubiquitin ligases, to demonstrate the feasibility of this strategy. Many potential substrates of LNX E3s were identified. Eight of the nine selected candidates were ubiquitinated in vitro, and two novel endogenous substrates, PDZ-binding kinase (PBK) and breakpoint cluster region protein (BCR), were confirmed in vivo. We further revealed that the LNX1-mediated ubiquitination and degradation of PBK inhibited cell proliferation and enhanced sensitivity to doxorubicin-induced apoptosis. The substrate recognition mechanism of LNX E3s was also characterized; this process involves the recognition of substrates via their specific PDZ domains by binding to the C-termini of the target proteins. This strategy can potentially be extended to a variety of E3s that contain protein interaction domain(s), thereby serving as a powerful tool for the comprehensive identification of their substrates on a proteomic scale.
Collapse
Affiliation(s)
- Zhengguang Guo
- National Key Laboratory of Medical Molecular Biology, Department of Physiology and Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences/School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Tao T, Cheng C, Ji Y, Xu G, Zhang J, Zhang L, Shen A. Numbl inhibits glioma cell migration and invasion by suppressing TRAF5-mediated NF-κB activation. Mol Biol Cell 2012; 23:2635-44. [PMID: 22593207 PMCID: PMC3395653 DOI: 10.1091/mbc.e11-09-0805] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Numblike, a negative regulator in glioma cell migration and invasion, was found to mediate nuclear factor kappa B activation by suppressing tumor necrosis factor receptor–associated factor 5. The Notch signaling regulator Numblike (Numbl) is expressed in the brain, but little is known regarding its role in the pathophysiology of glial cells. In this paper, we report that Numbl expression was down-regulated in high-grade human glioma tissue samples and glioblastoma cell lines. To investigate the role of Numbl in glioma migration and invasion, we generated human glioma cell lines in which Numbl was either overexpressed or depleted. Overexpression of Numbl suppressed, while elimination of Numbl promoted, the migration and invasion of glioma cells. Numbl inhibited glioma migration and invasion by dampening NF-κB activity. Furthermore, Numbl interacted directly with tumor necrosis factor receptor–associated factor 5 (TRAF5), which signals upstream and is required for the activation of NF-κB, and committed it to proteasomal degradation by promoting K48-linked polyubiquitination of TRAF5. In conclusion, our data suggest that Numbl negative regulates glioma cell migration and invasion by abrogating TRAF5-induced activation of NF-κB.
Collapse
Affiliation(s)
- Tao Tao
- Key Laboratory of Neuroregeneration of Jiangsu Province, Nantong University, Nantong, Jiangsu, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
10
|
Kresse SH, Meza-Zepeda LA, Machado I, Llombart-Bosch A, Myklebost O. Preclinical xenograft models of human sarcoma show nonrandom loss of aberrations. Cancer 2011; 118:558-70. [PMID: 21713766 DOI: 10.1002/cncr.26276] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 04/07/2011] [Accepted: 04/21/2011] [Indexed: 01/04/2023]
Abstract
BACKGROUND Human tumors transplanted into immunodeficient mice (xenografts) are good preclinical models, and it is important to identify possible systematic changes during establishment and passaging in mice. METHODS High-resolution microarray-based comparative genomic hybridization (array CGH) was used to investigate how well a series of sarcoma xenografts, including 9 patient/xenograft pairs and 8 early versus late xenograft passage pairs, represented the patient tumor from which they originated. RESULTS In all analyses, the xenografts were more similar to their tumor of origin than other xenografts of the same type. Most changes in aberration patterns were toward a more normal genome complement, and the increased aberrations observed were mostly toward more loss. In general, the changes were scattered over the genome, but some changes were significant in osteosarcomas. These were rather focused and consistent with amplifications frequent in patient samples, involving the genes platelet-derived growth factor receptor A (PDGFRA), cysteine-rich hydrophobic domain 2 (CHIC2), FIP-like 1 (FIP1L1), ligand of numb-protein X1 (LNX1), RAS-like family 11 member B (RASL11B), and sec1 family domain containing 2 (SCFD2), probably a sign of continued tumor progression. Some changes that disappeared may have been involved in host-stroma interactions or chemotherapy resistance, possibly because of the absence of selection in the mouse. CONCLUSIONS Direct xenografts reflected well the genomic patterns of their tumors of origin. The few significant aberrations that were lost during passaging in immune-defective mice may have been caused by the lack of selection in the new host, whereas aberrations that were gained appeared to be the result of general tumor progression rather than model-specific artifacts.
Collapse
Affiliation(s)
- Stine H Kresse
- Department of Tumor Biology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | | | | | | | | |
Collapse
|