LRP6 Promotes Invasion and Metastasis of Colorectal Cancer Through Cytoskeleton Dynamics

Total Page:16

File Type:pdf, Size:1020Kb

LRP6 Promotes Invasion and Metastasis of Colorectal Cancer Through Cytoskeleton Dynamics www.impactjournals.com/oncotarget/ Oncotarget, 2017, Vol. 8, (No. 65), pp: 109632-109645 Research Paper LRP6 promotes invasion and metastasis of colorectal cancer through cytoskeleton dynamics Qian Yao1,*, Yu An1,*, Wei Hou1, Ya-Nan Cao1, Meng-Fei Yao1, Ning-Ning Ma1, Lin Hou1, Hong Zhang2, Hai-Jing Liu1 and Bo Zhang1 1Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China 2Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China *These authors have contributed equally to this work Correspondence to: Hai-Jing Liu, email: [email protected] Bo Zhang, email: [email protected] Keywords: colorectal cancer; Wnt signaling; LRP6; metastasis; cytoskeleton Received: May 09, 2017 Accepted: October 28, 2017 Published: November 30, 2017 Copyright: Yao et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License 3.0 (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Low density lipoprotein (LDL) receptor-related protein-6 (LRP6) is an important co-receptor of Wnt pathway, which plays a predominant role in development and progression of colorectal cancer. Recently, dysregulation of LRP6 has proved to be involved in the progression of cancers, but its biological role and clinical significance in colorectal cancer remain unclear. In present study, we revealed that phosphorylation of LRP6 was aberrantly upregulated in colorectal carcinoma correlating with TNM or Dukes staging and worse prognosis. In addition, phosphorylated LRP6 was positively correlated with nuclear accumulation of β-catenin. Overexpression or activation of LRP6 could activate Wnt signaling and promote tumor cell migration in vitro. The activation of LRP6 could induce microtubule dynamics and actin remodeling, probably through regulation of microtubule-associated protein 1B (MAP1B), microtubule actin cross-linking factor 1 (MACF1) and Rho GTPase--RhoA and Rac1. The investigation suggests that LRP6 may be a potential prognostic marker and therapeutic target in the progression of colorectal cancers. INTRODUCTION and progression of colorectal cancers in past decades. Especially, nuclear translocation of β-catenin, a hallmark Colorectal cancer is the third most common of Wnt signaling activation, has proved to be a critical cancer and fourth leading cause of cancer-related deaths role in many aspects of cellular biological activities, such worldwide [1, 2] and there is potential for distant as growth, differentiation, EMT, as well as cytoskeleton metastasis following surgical excision of the primary remodeling [6, 7]. Nevertheless, Wnt signaling could be lesion [3]. Despite the introduction of new treatments, further enhanced by its secreted ligands even in APC- the 5-year survival rate for metastatic colorectal cancer mutated situation, suggesting that that hyperactivation of remains below 10% [4]. Hence, the underlying mechanism Wnt pathway may be contributed by other alterations in of metastasis, predictive biomarkers and therapeutic targets colorectal cancers [8-11]. for colorectal cancer are under extensive exploration. Loss Low-density lipoprotein (LDL) receptor-related of function mutation in APC (adenomatous polyposis coli) protein-6 (LRP6) is one of the co-receptors of Wnt tumor suppressor gene has been detected in about 85% pathway, which form a signalosome with Wnt ligand of sporadic colorectal cancers, which leads to activation and Frizzled receptor to activate downstream cascade. of Wnt signaling pathway [5]. In fact, activation of Wnt The phosphorylation of PPSPXS motifs of LRP6 could signaling has been clarified to be involved in initiation recruit other Wnt signaling components Disheveled (Dvl), www.impactjournals.com/oncotarget 109632 Oncotarget Axin and GSK3β to signalosome to amplify and stabilize Positive p-LRP6 was found in 92 of 115 cases (80.0%) of its phosphorylation, which can directly inhibit GSK3β advanced carcinoma (T3 and T4), while early carcinomas activity leading to multiple downstream cascades [12- (T1 and T2) showed positive staining in just 35.3% (24/68) 14]. Therefore, LRP6 represents an important regulatory (p<0.001, χ2=36.801). 76.6% (59/77) of cases with lymph node in transducing Wnt stimulation. Recent reports reveal node metastasis was positive p-LRP6, but only 48.8% that LRP6 contributed to tumor progression in colorectal (39/80) (p<0.001, χ2=12.995) without metastasis showed cancer and some other cancers [15-18]. However, the p-LRP6 expression. In addition, there was a positive mechanism about LRP6 in colorectal cancers is seldom linear correlation between p-LRP6 expression and TNM investigated. or Dukes staging (p<0.001). Kaplan-Meier single-factor In this study, we demonstrated that phosphorylated analysis and log-rank test demonstrated a statistically LRP6 was markedly up-regulate in colorectal carcinoma significant decrease in disease-free survival (DFS) in and indicated poor prognosis. The activation of LRP6 patients with positive staining of p-LRP6 (p=0.003) could enhance Wnt signaling and promote cell migration. (Figure 1C). However, evaluation of patients’ overall LRP6 governed the cytoskeleton stability and remodeling survival (OS) displayed a borderline significance between through microtubule-related protein: microtubule- positive and negative expression of p-LRP6 (p=0.058) associated protein 1B (MAP1B) and microtubule actin (Figure 1D). cross-linking factor 1 (MACF1) as well as Rho GTPases. Taken together, these data proved that the p-LRP6 was involved in Wnt signaling activation and correlated RESULTS with a more aggressive phenotype with higher metastatic potential and worse prognosis. Expression of Phosphorylated LRP6 is correlated with aggressive clinical behavior in Activation of LRP6 increases Wnt/β-catenin colorectal carcinoma signaling in colorectal cancer cells Immuno-staining of both LRP6 and phosphorylated To evaluate the role of LRP6 in activation of Wnt LRP6 (p-LRP6) were performed in colorectal carcinoma signaling, the plasmids LRP6-WT (wild-type LRP6), tissue. The expression of LRP6 was negative in most cases LRP6-DA (dominant active, reserving the intracellular (91.2%, 62/68) with few positive (8.8%, 6/68). However, domain (ICD) of LRP6, residues 1126-1613) and LRP6- the immunostaining of p-LRP6 was in a diffuse pattern DN (dominant negative, reserving the extracellular domain in cytoplasm with no apparent membranous expression. of LRP6, residues 1-1114) were constructed (Figure 2A). Comparing to non-neoplastic glandular cells (4.8%, With transfection of Lovo and HCT116 cells with above 1/21) and adenoma tissue (5.3%, 2/38), the expression plasmids, β-catenin immunofluorescence staining and of p-LRP6 was much more intense in carcinoma with luciferase assay were carried out. The results showed that higher positive level (62.3%, 114/183), which displayed nuclear staining of β-catenin was obviously enhanced the significant statistical difference (p<0.001) (Figure 1A, in LRP6-WT and LRP6-DA group with attenuated 1B). It was noteworthy that positive p-LRP6 was usually membrane expression comparing to control or LRP6-DN observed at the invasive front tumor zone in infiltrating (p<0.001) (Figure 2B, 2C). Expectedly, overexpression of nests of cancers (Figure 1A). LRP6-WT and LRP6-DA led to an activation of TCF/β- Wnt signaling plays a pivotal role in colorectal catenin reporter up to about 2-fold, as compared to the carcinogenesis and the nuclear accumulation of β-catenin vector control or LRP6-DN (p<0.001) (Figure 2D). The is widely accepted as the hallmark of Wnt signaling data demonstrated that activation of LRP6 could increase activation [19, 20]. Accordingly, the expressions of Wnt signaling activity in colorectal cancer cells. β-catenin was immunohistochemically evaluated. Positive nuclear expression of β-catenin was 74.3% (136/183) in Activation of LRP6 promotes migration of carcinoma, which was higher than adenoma tissue (55.3%, colorectal cancer cells 21/38) (p<0.05) (Supplementary Figure 1). At the same time, the cytoplasm expression of β-catenin was also found To determine whether up-regulation of LRP6 would more frequently in carcinoma cells (79.8%, 146/183) than influence cancer cell migration and invasion, Transwell adenoma (57.9%, 22/38) (p<0.05) (Supplementary Figure assays with or without Matrigel were performed. In 1A, 1B). Besides, a positive correlation between p-LRP6 migration assay, Lovo and HCT116 cells transfected and nuclear expression of β-catenin was established with LRP6-WT or LRP6-DA plasmids demonstrated (Supplementary Table 1). much higher motility potential than vector control group The correlations between p-LRP6 expression and or LRP6-DN group (p<0.001) (Figure 2E, 2F). As for clinicopathologic parameters such as age, gender, tumor invasion assay, we failed to found the statistical difference size, depth of invasion, lymph node metastasis, TNM and between each group, however, the results showed the Dukes staging were analyzed (Supplementary Table 2). tendency of promoted invasive potential in LRP6-DA and www.impactjournals.com/oncotarget 109633 Oncotarget LRP6-WT transfected cells (p=0.087) (Supplementary invasion, which arise the question about how LRP6 Figure 2). manage to control such aggressive behavior. It was widely Above evidence suggested the possible role of LRP6 agreed that migratory cancer cells undergo
Recommended publications
  • Cyclin D1/Cyclin-Dependent Kinase 4 Interacts with Filamin a and Affects the Migration and Invasion Potential of Breast Cancer Cells
    Published OnlineFirst February 28, 2010; DOI: 10.1158/0008-5472.CAN-08-1108 Tumor and Stem Cell Biology Cancer Research Cyclin D1/Cyclin-Dependent Kinase 4 Interacts with Filamin A and Affects the Migration and Invasion Potential of Breast Cancer Cells Zhijiu Zhong, Wen-Shuz Yeow, Chunhua Zou, Richard Wassell, Chenguang Wang, Richard G. Pestell, Judy N. Quong, and Andrew A. Quong Abstract Cyclin D1 belongs to a family of proteins that regulate progression through the G1-S phase of the cell cycle by binding to cyclin-dependent kinase (cdk)-4 to phosphorylate the retinoblastoma protein and release E2F transcription factors for progression through cell cycle. Several cancers, including breast, colon, and prostate, overexpress the cyclin D1 gene. However, the correlation of cyclin D1 overexpression with E2F target gene regulation or of cdk-dependent cyclin D1 activity with tumor development has not been identified. This suggests that the role of cyclin D1 in oncogenesis may be independent of its function as a cell cycle regulator. One such function is the role of cyclin D1 in cell adhesion and motility. Filamin A (FLNa), a member of the actin-binding filamin protein family, regulates signaling events involved in cell motility and invasion. FLNa has also been associated with a variety of cancers including lung cancer, prostate cancer, melanoma, human bladder cancer, and neuroblastoma. We hypothesized that elevated cyclin D1 facilitates motility in the invasive MDA-MB-231 breast cancer cell line. We show that MDA-MB-231 motility is affected by disturbing cyclin D1 levels or cyclin D1-cdk4/6 kinase activity.
    [Show full text]
  • Snapshot: Formins Christian Baarlink, Dominique Brandt, and Robert Grosse University of Marburg, Marburg 35032, Germany
    SnapShot: Formins Christian Baarlink, Dominique Brandt, and Robert Grosse University of Marburg, Marburg 35032, Germany Formin Regulators Localization Cellular Function Disease Association DIAPH1/DIA1 RhoA, RhoC Cell cortex, Polarized cell migration, microtubule stabilization, Autosomal-dominant nonsyndromic deafness (DFNA1), myeloproliferative (mDia1) phagocytic cup, phagocytosis, axon elongation defects, defects in T lymphocyte traffi cking and proliferation, tumor cell mitotic spindle invasion, defects in natural killer lymphocyte function DIAPH2 Cdc42 Kinetochore Stable microtubule attachment to kinetochore for Premature ovarian failure (mDia3) chromosome alignment DIAPH3 Rif, Cdc42, Filopodia, Filopodia formation, removing the nucleus from Increased chromosomal deletion of gene locus in metastatic tumors (mDia2) Rac, RhoB, endosomes erythroblast, endosome motility, microtubule DIP* stabilization FMNL1 (FRLα) Cdc42 Cell cortex, Phagocytosis, T cell polarity Overexpression is linked to leukemia and non-Hodgkin lymphoma microtubule- organizing center FMNL2/FRL3/ RhoC ND Cell motility Upregulated in metastatic colorectal cancer, chromosomal deletion is FHOD2 associated with mental retardation FMNL3/FRL2 Constituently Stress fi bers ND ND active DAAM1 Dishevelled Cell cortex Planar cell polarity ND DAAM2 ND ND ND Overexpressed in schizophrenia patients Human (Mouse) FHOD1 ROCK Stress fi bers Cell motility FHOD3 ND Nestin, sarcomere Organizing sarcomeres in striated muscle cells Single-nucleotide polymorphisms associated with type 1 diabetes
    [Show full text]
  • Identification of a WNT5A-Responsive Degradation Domain in the Kinesin
    G C A T T A C G G C A T genes Article Identification of a WNT5A-Responsive Degradation Domain in the Kinesin Superfamily Protein KIF26B Edith P. Karuna ID , Shannon S. Choi, Michael K. Scales, Jennie Hum, Michael Cohen, Fernando A. Fierro and Hsin-Yi Henry Ho * ID Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, CA 95616, USA; [email protected] (E.P.K.); [email protected] (S.S.C.); [email protected] (M.K.S.); [email protected] (J.H.); [email protected] (M.C.); ffi[email protected] (F.A.F.) * Correspondence: [email protected]; Tel.: +1-530-752-8857 Received: 19 February 2018; Accepted: 26 March 2018; Published: 5 April 2018 Abstract: Noncanonical WNT pathways function independently of the β-catenin transcriptional co-activator to regulate diverse morphogenetic and pathogenic processes. Recent studies showed that noncanonical WNTs, such as WNT5A, can signal the degradation of several downstream effectors, thereby modulating these effectors’ cellular activities. The protein domain(s) that mediates the WNT5A-dependent degradation response, however, has not been identified. By coupling protein mutagenesis experiments with a flow cytometry-based degradation reporter assay, we have defined a protein domain in the kinesin superfamily protein KIF26B that is essential for WNT5A-dependent degradation. We found that a human disease-causing KIF26B mutation located at a conserved amino acid within this domain compromises the ability of WNT5A to induce KIF26B degradation. Using pharmacological perturbation, we further uncovered a role of glycogen synthase kinase 3 (GSK3) in WNT5A regulation of KIF26B degradation.
    [Show full text]
  • Supplementary Figures
    Mena regulates the LINC complex to control actin–nuclear lamina associations, trans-nuclear membrane signalling and cancer gene expression Frederic Li Mow Chee!, Bruno Beernaert!, Alexander Loftus!, Yatendra Kumar", Billie G. C. Griffith!, Jimi C. Wills!, Ann P. Wheeler#, J. Douglas Armstrong$, Maddy Parsons%, Irene M. Leigh,(, Charlotte M. Proby&, Alex von Kriegsheim!, Wendy A. Bickmore", Margaret C. Frame,* & Adam Byron,* Supplementary Information Supplementary Figure 1 Supplementary Figure 2 Supplementary Figure 3 Supplementary Table 1 Supplementary Table 2 Supplementary Table 3 Supplementary Table 4 !Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH< =XR, UK. "MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH< =XU, UK. #Advanced Imaging Resource, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH< =XU, UK. $Simons Initiative for the Developing Brain, School of Informatics, University of Edinburgh, Edinburgh EHH IYL, UK. %Randall Centre for Cell and Molecular Biophysics, King’s College London, London SEM MUL, UK. &Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee DD <HN, UK. 'Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EM =AT, UK. *email: [email protected] or [email protected] 1 a cSCC IAC correlation b cSCC IAC pathways c Core adhesome network ENAH −log10(q) MACF1 CSRP1 Met1 Met4 0 5 10 + + CORO2A Integrin signalling + CFL1 pathway PRNP ILK + HSPB1 PALLD PPFIA1 TES RDX Cytoskeletal regulation + VASP + + ARPC2 by Rho GTPase PPP2CA + Met1 + LASP1 MYH9 + VIM TUBA4A Huntington ITGA3 + disease ITGB4 VCL CAV1 ACTB ROCK1 KTN1 FLNA+ CALR DNA FBLIM1 CORO1B RAC1 + replication +ACTN1 ITGA6 + Met4 ITGAV Parkinson ITGB1 disease Actin cytoskel.
    [Show full text]
  • Universidade Estadual De Campinas Instituto De Biologia
    UNIVERSIDADE ESTADUAL DE CAMPINAS INSTITUTO DE BIOLOGIA VERÔNICA APARECIDA MONTEIRO SAIA CEREDA O PROTEOMA DO CORPO CALOSO DA ESQUIZOFRENIA THE PROTEOME OF THE CORPUS CALLOSUM IN SCHIZOPHRENIA CAMPINAS 2016 1 VERÔNICA APARECIDA MONTEIRO SAIA CEREDA O PROTEOMA DO CORPO CALOSO DA ESQUIZOFRENIA THE PROTEOME OF THE CORPUS CALLOSUM IN SCHIZOPHRENIA Dissertação apresentada ao Instituto de Biologia da Universidade Estadual de Campinas como parte dos requisitos exigidos para a obtenção do Título de Mestra em Biologia Funcional e Molecular na área de concentração de Bioquímica. Dissertation presented to the Institute of Biology of the University of Campinas in partial fulfillment of the requirements for the degree of Master in Functional and Molecular Biology, in the area of Biochemistry. ESTE ARQUIVO DIGITAL CORRESPONDE À VERSÃO FINAL DA DISSERTAÇÃO DEFENDIDA PELA ALUNA VERÔNICA APARECIDA MONTEIRO SAIA CEREDA E ORIENTADA PELO DANIEL MARTINS-DE-SOUZA. Orientador: Daniel Martins-de-Souza CAMPINAS 2016 2 Agência(s) de fomento e nº(s) de processo(s): CNPq, 151787/2F2014-0 Ficha catalográfica Universidade Estadual de Campinas Biblioteca do Instituto de Biologia Mara Janaina de Oliveira - CRB 8/6972 Saia-Cereda, Verônica Aparecida Monteiro, 1988- Sa21p O proteoma do corpo caloso da esquizofrenia / Verônica Aparecida Monteiro Saia Cereda. – Campinas, SP : [s.n.], 2016. Orientador: Daniel Martins de Souza. Dissertação (mestrado) – Universidade Estadual de Campinas, Instituto de Biologia. 1. Esquizofrenia. 2. Espectrometria de massas. 3. Corpo caloso.
    [Show full text]
  • Plakins, a Versatile Family of Cytolinkers: Roles in Skin Integrity and in Human Diseases Jamal-Eddine Bouameur1,2, Bertrand Favre1 and Luca Borradori1
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Elsevier - Publisher Connector REVIEW Plakins, a Versatile Family of Cytolinkers: Roles in Skin Integrity and in Human Diseases Jamal-Eddine Bouameur1,2, Bertrand Favre1 and Luca Borradori1 The plakin family consists of giant proteins involved in in (Roper et al., 2002; Jefferson et al., 2004; Sonnenberg and the cross-linking and organization of the cytoskeleton Liem, 2007; Boyer et al., 2010; Suozzi et al., 2012). and adhesion complexes. They further modulate sev- Mammalian plakins share a similar structural organization eral fundamental biological processes, such as cell and comprise seven members: bullous pemphigoid antigen 1 adhesion, migration, and polarization or signaling (BPAG1), desmoplakin, envoplakin, epiplakin, microtubule- pathways. Inherited and acquired defects of plakins actin cross-linking factor 1 (MACF1), periplakin, and plectin in humans and in animal models potentially lead to (Figure 1) (Choi et al., 2002; Jefferson et al., 2007; Choi and dramatic manifestations in the skin, striated muscles, Weis, 2011; Ortega et al., 2011). The existence of develop- and/or nervous system. These observations unequivo- mentally regulated and tissue-specific splice variants of some cally demonstrate the key role of plakins in the plakins further increases the diversity and versatility of these proteins (Table 1; Figure 1; Leung et al., 2001; Rezniczek et al., maintenance of tissue integrity. Here we review the 2003; Lin et al., 2005; Jefferson et al., 2006; Cabral et al., 2010). characteristics of the mammalian plakin members BPAG1 (bullous pemphigoid antigen 1), desmoplakin, PLAKINS IN THE EPIDERMIS plectin, envoplakin, epiplakin, MACF1 (microtubule- Epithelial BPAG1 (BPAG1e, also called BP230) constitutes the actin cross-linking factor 1), and periplakin, highlight- epithelium-specific isoform of BPAG1 and is localized in basal ing their role in skin homeostasis and diseases.
    [Show full text]
  • How Microtubules Control Focal Adhesion Dynamics
    JCB: Review Targeting and transport: How microtubules control focal adhesion dynamics Samantha Stehbens and Torsten Wittmann Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143 Directional cell migration requires force generation that of integrin-mediated, nascent adhesions near the cell’s leading relies on the coordinated remodeling of interactions with edge, which either rapidly turn over or connect to the actin cytoskeleton (Parsons et al., 2010). Actomyosin-mediated the extracellular matrix (ECM), which is mediated by pulling forces allow a subset of these nascent FAs to grow integrin-based focal adhesions (FAs). Normal FA turn- and mature, and provide forward traction forces. However, in over requires dynamic microtubules, and three members order for cells to productively move forward, FAs also have to of the diverse group of microtubule plus-end-tracking release and disassemble underneath the cell body and in the proteins are principally involved in mediating micro- rear of the cell. Spatial and temporal control of turnover of tubule interactions with FAs. Microtubules also alter these mature FAs is important, as they provide a counterbalance to forward traction forces, and regulated FA disassembly is the assembly state of FAs by modulating Rho GTPase required for forward translocation of the cell body. An important signaling, and recent evidence suggests that microtubule- question that we are only beginning to understand is how FA mediated clathrin-dependent and -independent endo­ turnover is spatially and temporally regulated to allow cells cytosis regulates FA dynamics. In addition, FA-associated to appropriately respond to extracellular signals, allowing for microtubules may provide a polarized microtubule track for coordinated and productive movement.
    [Show full text]
  • Isoforms, Structures, and Functions of Versatile Spectraplakin MACF1
    BMB Rep. 2016; 49(1): 37-44 BMB www.bmbreports.org Reports Contiributed Mini Review Isoforms, structures, and functions of versatile spectraplakin MACF1 Lifang Hu1, Peihong Su1, Runzhi Li1, Chong Yin1, Yan Zhang1, Peng Shang1, Tuanmin Yang2 & Airong Qian1,* 1Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi 710072, 2Honghui Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shaanxi 710054, P. R. China Spectraplakins are crucially important communicators, linking ability of interacting with all three types of cytoskeletal fila- cytoskeletal components to each other and cellular junctions. ments, i.e., F-actin, MTs, and IFs (1). Microtubule actin crosslinking factor 1 (MACF1), also known Spectraplakins are exceptionally long and gigantic with mo- as actin crosslinking family 7 (ACF7), is a member of the spec- lecular weight of >500 kD. They are multi-domain cytoskele- traplakin family. It is expressed in numerous tissues and cells tal proteins and master orchestrators for coordinating cytoske- as one extensively studied spectraplakin. MACF1 has several letal elements by binding to F-actin, MTs, and IFs (1, 2). isoforms with unique structures and well-known function to be Spectraplakins are evolutionarily conserved. They belong to able to crosslink F-actin and microtubules. MACF1 is one ver- both spectrin and plakin superfamilies (3). “Spectraplakins” are satile spectraplakin with various functions in cell processes, named as combinations of “spectrin” and “plakin” because embryo development, tissue-specific functions, and human they share features of both spectrins and plakins (3, 4). So far, diseases. The importance of MACF1 has become more appa- mammalian spectraplakins have two members: microtubule rent in recent years.
    [Show full text]
  • Harnessing Low-Density Lipoprotein Receptor Protein 6 (LRP6) Genetic Variation and Wnt Signaling for Innovative Diagnostics in Complex Diseases
    OPEN The Pharmacogenomics Journal (2018) 18, 351–358 www.nature.com/tpj REVIEW Harnessing low-density lipoprotein receptor protein 6 (LRP6) genetic variation and Wnt signaling for innovative diagnostics in complex diseases Z-M Wang1,2, J-Q Luo1,2, L-Y Xu3, H-H Zhou1,2 and W Zhang1,2 Wnt signaling regulates a broad variety of processes in both embryonic development and various diseases. Recent studies indicated that some genetic variants in Wnt signaling pathway may serve as predictors of diseases. Low-density lipoprotein receptor protein 6 (LRP6) is a Wnt co-receptor with essential functions in the Wnt/β-catenin pathway, and mutations in LRP6 gene are linked to many complex human diseases, including metabolic syndrome, cancer, Alzheimer’s disease and osteoporosis. Therefore, we focus on the role of LRP6 genetic polymorphisms and Wnt signaling in complex diseases, and the mechanisms from mouse models and cell lines. It is also highly anticipated that LRP6 variants will be applied clinically in the future. The brief review provided here could be a useful resource for future research and may contribute to a more accurate diagnosis in complex diseases. The Pharmacogenomics Journal (2018) 18, 351–358; doi:10.1038/tpj.2017.28; published online 11 July 2017 INTRODUCTION signaling pathways and expressed in various target organs.1 LDLR- The Wnt1 gene was identified in 1982. Ensuing studies in related proteins 5/6 (LRP5/6) belong to this large family and Drosophila and Xenopus unveiled a highly conserved Wnt/ function as co-receptors of the Wnt/β-catenin pathway. These β-catenin pathway, namely, canonical Wnt signaling.
    [Show full text]
  • The LRP6 Rs2302685 Polymorphism Is Associated with Increased Risk Of
    Xu et al. Lipids in Health and Disease 2014, 13:94 http://www.lipidworld.com/content/13/1/94 RESEARCH Open Access The LRP6 rs2302685 polymorphism is associated with increased risk of myocardial infarction Shun Xu1,2,3, Jie Cheng1,2,3, Yu-ning Chen1,2,3, Keshen Li4, Ze-wei Ma1,2, Jin-ming Cen5, Xinguang Liu1,2,3, Xi-li Yang5, Can Chen6 and Xing-dong Xiong1,2,3* Abstract Background: Abnormal lipids is one of the critical risk factors for myocardial infarction (MI), however the role of genetic variants in lipid metabolism-related genes on MI pathogenesis still requires further investigation. We herein genotyped three SNPs (LRP6 rs2302685, LDLRAP1 rs6687605, SOAT1 rs13306731) in lipid metabolism-related genes, aimed to shed light on the influence of these SNPs on individual susceptibility to MI. Methods: Genotyping of the three SNPs (rs2302685, rs6687605 and rs13306731) was performed in 285 MI cases and 650 control subjects using polymerase chain reaction–ligation detection reaction (PCR–LDR) method. The association of these SNPs with MI and lipid profiles was performed with SPSS software. Results: Multivariate logistic regression analysis showed that C allele (OR = 1.62, P = 0.039) and the combined CT/CC genotype (OR = 1.67, P = 0.035) of LRP6 rs2302685 were associated with increased MI risk, while the other two SNPs had no significant effect. Further stratified analysis uncovered a more evident association with MI risk among younger subjects (≤60 years old). Fascinatingly, CT/CC genotype of rs2302685 conferred increased LDL-C levels compared to TT genotype (3.0 mmol/L vs 2.72 mmol/L) in younger subjects.
    [Show full text]
  • A Global Transcriptional Network Connecting Noncoding Mutations to Changes in Tumor Gene Expression
    HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Nat Genet Manuscript Author . Author manuscript; Manuscript Author available in PMC 2018 October 02. Published in final edited form as: Nat Genet. 2018 April ; 50(4): 613–620. doi:10.1038/s41588-018-0091-2. A global transcriptional network connecting noncoding mutations to changes in tumor gene expression Wei Zhang1,†,*, Ana Bojorquez-Gomez1,†, Daniel Ortiz Velez2, Guorong Xu3, Kyle S. Sanchez1, John Paul Shen1, Kevin Chen2, Katherine Licon1, Collin Melton4, Katrina M. Olson1,5, Michael Ku Yu1, Justin K. Huang1,6, Hannah Carter1, Emma K. Farley1,5, Michael Snyder4, Stephanie I. Fraley2, Jason F. Kreisberg1,*, and Trey Ideker1,2,6,* 1Department of Medicine, University of California, San Diego, La Jolla, California, USA 2Department of Bioengineering, University of California San Diego, La Jolla, CA, USA 3Center for Computational Biology and Bioinformatics, University of California, San Diego, La Jolla, California, USA 4Department of Genetics, Stanford University School of Medicine, Stanford, California, USA 5Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA 6Bioinformatics and Systems Biology Program, UC San Diego, La Jolla, California, USA Abstract Although cancer genomes are replete with noncoding mutations, the effects of these mutations remain poorly characterized. Here we perform an integrative analysis of 930 tumor whole genomes and matched transcriptomes, identifying a network of 193 noncoding loci in which mutations disrupt target gene expression. These “somatic eQTLs” (expression Quantitative Trait Loci) are frequently mutated in specific cancer tissues, and the majority can be validated in an independent cohort of 3,382 tumors. Among these, we find that the effects of noncoding mutations on DAAM1, MTG2 and HYI transcription are recapitulated in multiple cancer cell lines, and that increasing DAAM1 expression leads to invasive cell migration.
    [Show full text]
  • Daam2 Couples Translocation and Clustering of Wnt Receptor Signalosomes Through Rac1 Carlo D
    © 2021. Published by The Company of Biologists Ltd | Journal of Cell Science (2021) 134, jcs251140. doi:10.1242/jcs.251140 RESEARCH ARTICLE Daam2 couples translocation and clustering of Wnt receptor signalosomes through Rac1 Carlo D. Cristobal1,QiYe2, Juyeon Jo2, Xiaoyun Ding3, Chih-Yen Wang2, Diego Cortes2, Zheng Chen4 and Hyun Kyoung Lee1,3,5,* ABSTRACT Dynamic polymerization of the Dishevelled proteins functions at Wnt signaling plays a critical role in development across species and the core of the Wnt signalosome by interacting with both the is dysregulated in a host of human diseases. A key step in signal Frizzled Wnt receptors and low-density lipoprotein receptor-related transduction is the formation of Wnt receptor signalosomes, during protein 5/6 (LRP5/6), leading to recruitment of Axin proteins β which a large number of components translocate to the membrane, from the -catenin destruction complex (MacDonald et al., 2009; cluster together and amplify downstream signaling. However, the Schwarz-Romond et al., 2007). However, the exact composition molecular processes that coordinate these events remain poorly and mechanisms of signalosome assembly at the plasma membrane defined. Here, we show that Daam2 regulates canonical Wnt remain unclear. signaling via the PIP –PIP5K axis through its association with Rac1. The hallmark of canonical Wnt signaling is the accumulation and 2 β Clustering of Daam2-mediated Wnt receptor complexes requires both translocation of -catenin into the nucleus for gene transcription, β Rac1 and PIP5K, and PIP5K promotes membrane localization of these whereas non-canonical Wnt signaling is -catenin independent and complexes in a Rac1-dependent manner. Importantly, the localization involves assembly/disassembly of the actin cytoskeleton, polarized of Daam2 complexes and Daam2-mediated canonical Wnt signaling is cell shape changes and cell migration (Niehrs, 2012; Schlessinger dependent upon actin polymerization.
    [Show full text]