1
|
Xiong X, Lee HC, Lu T. Impact of Sorbs2 dysfunction on cardiovascular diseases. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167813. [PMID: 40139410 PMCID: PMC12037213 DOI: 10.1016/j.bbadis.2025.167813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
Despite significant advancements in prevention and treatment over the past decades, cardiovascular diseases (CVDs) remain the leading cause of death worldwide. CVDs involve multifactorial inheritance, but our understanding of the genetic impact on these diseases is still incomplete. Sorbin and SH3 domain-containing protein 2 (Sorbs2) is ubiquitously expressed in various tissues, including the cardiovascular system. Increasing evidence suggests that Sorbs2 malfunction contributes to CVDs. This manuscript will review our current understanding of the potential mechanisms underlying Sorbs2 dysregulation in the development of CVDs.
Collapse
Affiliation(s)
- Xiaowei Xiong
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Hon-Chi Lee
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Tong Lu
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America.
| |
Collapse
|
2
|
Souza CB, Menezes GRO, Gondo A, Egito AA, Ramos PVB, Gomes RC, Ribas MN, Fernandes Júnior JA, Guimarães SEF. Estimation of Genetic Parameters and GWAS on Water Efficiency Traits in the Senepol Cattle. J Anim Breed Genet 2024. [PMID: 39726399 DOI: 10.1111/jbg.12920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/20/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024]
Abstract
The need for producing in environmentally resilient system drives new research to achieve sustainable beef production. Water footprint of the beef supply chain is a concern that must be addressed, aiming to improve water use within the production chain. One approach is genetic selection of beef cattle for water efficiency. However, it is essential to understand the genetic architecture and mechanisms involved in the expression of this phenotype to choose the best selection criteria. Thus, our study aimed to estimate genetic parameters for water efficiency traits, conduct a genome-wide association study (GWAS) and identify the genetic networks and biological processes involved. A population of 1762 purebred Senepol cattle was phenotyped for the following water efficiency traits: water intake (WI), gross water efficiency (GWE), water conversion ratio (WCR), residual water intake based on average daily gain (RWIADG) and residual water intake based on dry matter intake (RWIDMI). A subset of 1342 animals was genotyped using GGP Bovine 50 K SNP Chip with (734 animals) or 100 K (508 animals), and imputation from 50 K to 100 K was performed with Beagle software. The heritability estimates were 0.36 ± 0.06, 0.26 ± 0.05, 0.22 ± 0.05, 0.24 ± 0.05 and 0.20 ± 0.05 for WI, GWE, WCR, RWIADG and RWIDMI, respectively. Unlike the raw measures of WI, the phenotypic correlations between average daily gain (ADG) and the residuals (RWIDMI and RWIADG) were zero. All water efficiency traits were moderately to highly correlated with each other. GWAS were used to estimate the effect of 79,860 single nucleotide polymorphisms (SNPs), and significant SNPs were only observed for WCR. Enrichment analysis of genes in the significant regions revealed the involvement of different biological processes, such as saliva production, water transport, renal system and immune system. Genetic selection of Senepol cattle for water efficiency traits is feasible and can reduce water requirements for meat production. Water efficiency measures are polygenic traits, and different biological processes act simultaneously on the expression of related phenotypes.
Collapse
Affiliation(s)
- Christhian B Souza
- Departament of Animal Science, Federal University of Viçosa, Viçosa, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Abstract
BACKGROUND Vascular large conductance Ca2+-activated K+ (BK) channel, composed of the α-subunit (BK-α) and the β1-subunit (BK-β1), is a key determinant of coronary vasorelaxation and its function is impaired in diabetic vessels. However, our knowledge of diabetic BK channel dysregulation is incomplete. The Sorbs2 (Sorbin homology [SoHo] and Src homology 3 [SH3] domains-containing protein 2), is ubiquitously expressed in arteries, but its role in vascular pathophysiology is unknown. METHODS The role of Sorbs2 in regulating vascular BK channel activity was determined using patch-clamp recordings, molecular biological techniques, and in silico analysis. RESULTS Sorbs2 is not only a cytoskeletal protein but also an RNA-binding protein that binds to BK channel proteins and BK-α mRNA, regulating BK channel expression and function in coronary smooth muscle cells. Molecular biological studies reveal that the SH3 domain of Sorbs2 is necessary for Sorbs2 interaction with BK-α subunits, while both the SH3 and SoHo domains of Sorbs2 interact with BK-β1 subunits. Deletion of the SH3 or SoHo domains abolishes the Sorbs2 effect on the BK-α/BK-β1 channel current density. Additionally, Sorbs2 is a target gene of the Nrf2 (nuclear factor erythroid-2-related factor 2), which binds to the promoter of Sorbs2 and regulates Sorbs2 expression in coronary smooth muscle cells. In vivo studies demonstrate that Sorbs2 knockout mice at 4 months of age display a significant decrease in BK channel expression and function, accompanied by impaired BK channel Ca2+-sensitivity and BK channel-mediated vasodilation in coronary arteries, without altering their body weights and blood glucose levels. Importantly, Sorbs2 expression is significantly downregulated in the coronary arteries of db/db type 2 diabetic mice. CONCLUSIONS Sorbs2, a downstream target of Nrf2, plays an important role in regulating BK channel expression and function in vascular smooth muscle cells. Vascular Sorbs2 is downregulated in diabetes. Genetic knockout of Sorbs2 manifests coronary BK channelopathy and vasculopathy observed in diabetic mice, independent of obesity and glucotoxicity.
Collapse
Affiliation(s)
- Xiaojing Sun
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Hon-Chi Lee
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Tong Lu
- The Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
4
|
Lizcano-Perret B, Vertommen D, Herinckx G, Calabrese V, Gatto L, Roux PP, Michiels T. Identification of RSK substrates using an analog-sensitive kinase approach. J Biol Chem 2024; 300:105739. [PMID: 38342435 PMCID: PMC10945272 DOI: 10.1016/j.jbc.2024.105739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/28/2024] [Accepted: 02/02/2024] [Indexed: 02/13/2024] Open
Abstract
The p90 ribosomal S6 kinases (RSK) family of serine/threonine kinases comprises four isoforms (RSK1-4) that lie downstream of the ERK1/2 mitogen-activated protein kinase pathway. RSKs are implicated in fine tuning of cellular processes such as translation, transcription, proliferation, and motility. Previous work showed that pathogens such as Cardioviruses could hijack any of the four RSK isoforms to inhibit PKR activation or to disrupt cellular nucleocytoplasmic trafficking. In contrast, some reports suggest nonredundant functions for distinct RSK isoforms, whereas Coffin-Lowry syndrome has only been associated with mutations in the gene encoding RSK2. In this work, we used the analog-sensitive kinase strategy to ask whether the cellular substrates of distinct RSK isoforms differ. We compared the substrates of two of the most distant RSK isoforms: RSK1 and RSK4. We identified a series of potential substrates for both RSKs in cells and validated RanBP3, PDCD4, IRS2, and ZC3H11A as substrates of both RSK1 and RSK4, and SORBS2 as an RSK1 substrate. In addition, using mutagenesis and inhibitors, we confirmed analog-sensitive kinase data showing that endogenous RSKs phosphorylate TRIM33 at S1119. Our data thus identify a series of potential RSK substrates and suggest that the substrates of RSK1 and RSK4 largely overlap and that the specificity of the various RSK isoforms likely depends on their cell- or tissue-specific expression pattern.
Collapse
Affiliation(s)
- Belén Lizcano-Perret
- Molecular Virology Unit, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Didier Vertommen
- MASSPROT Platform, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Gaëtan Herinckx
- MASSPROT Platform, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Viviane Calabrese
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, Canada
| | - Laurent Gatto
- Computational Biology and Bioinformatics Unit, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Philippe P Roux
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, Canada; Faculty of Medicine, Department of Pathology and Cell Biology, Université de Montréal, Montreal, Quebec, Canada
| | - Thomas Michiels
- Molecular Virology Unit, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
5
|
Dan Y, Radic N, Gay M, Fernández-Torras A, Arauz G, Vilaseca M, Aloy P, Canovas B, Nebreda A. Characterization of p38α signaling networks in cancer cells using quantitative proteomics and phosphoproteomics. Mol Cell Proteomics 2023; 22:100527. [PMID: 36894123 PMCID: PMC10105487 DOI: 10.1016/j.mcpro.2023.100527] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
p38α (encoded by MAPK14) is a protein kinase that regulates cellular responses to almost all types of environmental and intracellular stresses. Upon activation, p38α phosphorylates many substrates both in the cytoplasm and nucleus, allowing this pathway to regulate a wide variety of cellular processes. While the role of p38α in the stress response has been widely investigated, its implication in cell homeostasis is less understood. To investigate the signaling networks regulated by p38α in proliferating cancer cells, we performed quantitative proteomic and phosphoproteomic analyses in breast cancer cells in which this pathway had been either genetically targeted or chemically inhibited. Our study identified with high confidence 35 proteins and 82 phosphoproteins (114 phosphosites) that are modulated by p38α, and highlighted the implication of various protein kinases, including MK2 and mTOR, in the p38α-regulated signaling networks. Moreover, functional analyses revealed an important contribution of p38α to the regulation of cell adhesion, DNA replication and RNA metabolism. Indeed, we provide experimental evidence supporting that p38α facilitates cancer cell adhesion, and showed that this p38α function is likely mediated by the modulation of the adaptor protein ArgBP2. Collectively, our results illustrate the complexity of the p38α regulated signaling networks, provide valuable information on p38α-dependent phosphorylation events in cancer cells, and document a mechanism by which p38α can regulate cell adhesion.
Collapse
Affiliation(s)
- Yuzhen Dan
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Nevenka Radic
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Marina Gay
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Adrià Fernández-Torras
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Gianluca Arauz
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Marta Vilaseca
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Patrick Aloy
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| | - Begoña Canovas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - AngelR Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain.
| |
Collapse
|
6
|
Maddala R, Eldawy C, Bachman W, Soderblom EJ, Rao PV. Glypican-4 regulated actin cytoskeletal reorganization in glucocorticoid treated trabecular meshwork cells and involvement of Wnt/PCP signaling. J Cell Physiol 2023; 238:631-646. [PMID: 36727620 DOI: 10.1002/jcp.30953] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/30/2022] [Accepted: 01/10/2023] [Indexed: 02/03/2023]
Abstract
A common adverse response to the clinical use of glucocorticoids (GCs) is elevated intraocular pressure (IOP) which is a major risk factor for glaucoma. Elevated IOP arises due to impaired outflow of aqueous humor (AH) through the trabecular meshwork (TM). Although GC-induced changes in actin cytoskeletal dynamics, contractile characteristics, and cell adhesive interactions of TM cells are believed to influence AH outflow and IOP, the molecular mechanisms mediating changes in these cellular characteristics are poorly understood. Our studies focused on evaluating changes in the cytoskeletal and cytoskeletal-associated protein (cytoskeletome) profile of human TM cells treated with dexamethasone (Dex) using label-free mass spectrometric quantification, identified elevated levels of specific proteins known to regulate actin stress fiber formation, contraction, actin networks crosslinking, cell adhesion, and Wnt signaling, including LIMCH1, ArgBP2, CNN3, ITGBL1, CTGF, palladin, FAT1, DIAPH2, EPHA4, SIPA1L1, and GPC4. Several of these proteins colocalized with the actin cytoskeleton and underwent alterations in distribution profile in TM cells treated with Dex, and an inhibitor of Abl/Src kinases. Wnt/Planar Cell Polarity (PCP) signaling agonists-Wnt5a and 5b were detected prominently in the cytoskeletome fraction of TM cells, and studies using siRNA to suppress expression of glypican-4 (GPC4), a known modulator of the Wnt/PCP pathway revealed that GPC4 deficiency impairs Dex induced actin stress fiber formation, and activation of c-Jun N-terminal Kinase (JNK) and Rho kinase. Additionally, while Dex augmented, GPC4 deficiency suppressed the formation of actin stress fibers in TM cells in the presence of Dex and Wnt5a. Taken together, these results identify the GPC4-dependent Wnt/PCP signaling pathway as one of the crucial upstream regulators of Dex induced actin cytoskeletal reorganization and cell adhesion in TM cells, opening an opportunity to target the GPC4/Wnt/PCP pathway for treatment of ocular hypertension in glaucoma.
Collapse
Affiliation(s)
- Rupalatha Maddala
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Camelia Eldawy
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - William Bachman
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Erik J Soderblom
- Proteomics and Metabolomics Shared Resource, Duke University School of Medicine, Duke Center for Genomic and Computational Biology, Durham, North Carolina, USA
| | - Ponugoti V Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
7
|
Biomolecular condensate assembly of nArgBP2 tunes its functionality to manifest the structural plasticity of dendritic spines. Exp Mol Med 2023; 55:108-119. [PMID: 36599935 PMCID: PMC9898538 DOI: 10.1038/s12276-022-00918-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 01/05/2023] Open
Abstract
nArgBP2, a candidate gene for intellectual disability, is a postsynaptic protein critical for dendritic spine development and morphogenesis, and its knockdown (KD) in developing neurons severely impairs spine-bearing excitatory synapse formation. Surprisingly, nArgBP2 KD in mature neurons did not cause morphological defects in the existing spines at rest, raising questions of how it functions in mature neurons. We found that unlike its inaction at rest, nArgBP2 KD completely inhibited the enlargement of dendritic spines during chemically induced long-term potentiation (cLTP) in mature neurons. We further found that nArgBP2 forms condensates in dendritic spines and that these condensates are dispersed by cLTP, which spatiotemporally coincides with spine head enlargement. Condensates with CaMKII phosphorylation-deficient mutant or CaMKII inhibition are neither dispersed nor accompanied by spine enlargement during cLTP. We found that nArgBP2 condensates in spines exhibited liquid-like properties, and in heterologous and in vitro expression systems, nArgBP2 undergoes liquid-liquid phase separation via multivalent intermolecular interactions between SH3 domains and proline-rich domains. It also forms coacervates with CaMKIIα, which is rapidly dissembled by calcium/CaMKIIα-dependent phosphorylation. We further showed that the interaction between nArgBP2 and WAVE1 competes with nArgBP2 phase separation and that blocking the nArgBP2-WAVE1 interaction prevents spine enlargement during cLTP. Together, our results suggest that nArgBP2 at rest is confined to the condensates but is released by CaMKIIα-mediated phosphorylation during synaptic plasticity, which regulates its timely interaction with WAVE1 to induce spine head enlargement in mature neurons.
Collapse
|
8
|
Advances in the previous two decades in our understanding of the post-translational modifications, functions, and drug perspectives of ArgBP2 and its family members. Biomed Pharmacother 2022; 155:113853. [DOI: 10.1016/j.biopha.2022.113853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 11/20/2022] Open
|
9
|
Svec KV, Howe AK. Protein Kinase A in cellular migration-Niche signaling of a ubiquitous kinase. Front Mol Biosci 2022; 9:953093. [PMID: 35959460 PMCID: PMC9361040 DOI: 10.3389/fmolb.2022.953093] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/30/2022] [Indexed: 12/28/2022] Open
Abstract
Cell migration requires establishment and maintenance of directional polarity, which in turn requires spatial heterogeneity in the regulation of protrusion, retraction, and adhesion. Thus, the signaling proteins that regulate these various structural processes must also be distinctly regulated in subcellular space. Protein Kinase A (PKA) is a ubiquitous serine/threonine kinase involved in innumerable cellular processes. In the context of cell migration, it has a paradoxical role in that global inhibition or activation of PKA inhibits migration. It follows, then, that the subcellular regulation of PKA is key to bringing its proper permissive and restrictive functions to the correct parts of the cell. Proper subcellular regulation of PKA controls not only when and where it is active but also specifies the targets for that activity, allowing the cell to use a single, promiscuous kinase to exert distinct functions within different subcellular niches to facilitate cell movement. In this way, understanding PKA signaling in migration is a study in context and in the elegant coordination of distinct functions of a single protein in a complex cellular process.
Collapse
Affiliation(s)
- Kathryn V. Svec
- Department of Pharmacology, University of Vermont, Burlington, VT, United States
| | - Alan K. Howe
- Department of Pharmacology, University of Vermont, Burlington, VT, United States
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, V T, United States
- University of Vermont Cancer Center, University of Vermont, Burlington, VT, United States
| |
Collapse
|
10
|
Wu H, Gonzalez Villalobos R, Yao X, Reilly D, Chen T, Rankin M, Myshkin E, Breyer MD, Humphreys BD. Mapping the single-cell transcriptomic response of murine diabetic kidney disease to therapies. Cell Metab 2022; 34:1064-1078.e6. [PMID: 35709763 PMCID: PMC9262852 DOI: 10.1016/j.cmet.2022.05.010] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 03/21/2022] [Accepted: 05/24/2022] [Indexed: 11/29/2022]
Abstract
Diabetic kidney disease (DKD) occurs in ∼40% of patients with diabetes and causes kidney failure, cardiovascular disease, and premature death. We analyzed the response of a murine DKD model to five treatment regimens using single-cell RNA sequencing (scRNA-seq). Our atlas of ∼1 million cells revealed a heterogeneous response of all kidney cell types both to DKD and its treatment. Both monotherapy and combination therapies targeted differing cell types and induced distinct and non-overlapping transcriptional changes. The early effects of sodium-glucose cotransporter-2 inhibitors (SGLT2i) on the S1 segment of the proximal tubule suggest that this drug class induces fasting mimicry and hypoxia responses. Diabetes downregulated the spliceosome regulator serine/arginine-rich splicing factor 7 (Srsf7) in proximal tubule that was specifically rescued by SGLT2i. In vitro proximal tubule knockdown of Srsf7 induced a pro-inflammatory phenotype, implicating alternative splicing as a driver of DKD and suggesting SGLT2i regulation of proximal tubule alternative splicing as a potential mechanism of action for this drug class.
Collapse
Affiliation(s)
- Haojia Wu
- Division of Nephrology, Department of Medicine, Washington University, St. Louis, MO, USA
| | | | - Xiang Yao
- Tox LJ Janssen Research & Development, La Jolla, CA, USA
| | | | - Tao Chen
- PSTS Janssen Research & Development, Shanghai, China
| | | | | | | | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University, St. Louis, MO, USA; Department of Developmental Biology, Washington University, St. Louis, MO, USA.
| |
Collapse
|
11
|
Morris T, Sue E, Geniesse C, Brieher WM, Tang VW. Synaptopodin stress fiber and contractomere at the epithelial junction. J Cell Biol 2022; 221:e202011162. [PMID: 35416930 PMCID: PMC9011326 DOI: 10.1083/jcb.202011162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 10/07/2021] [Accepted: 02/09/2022] [Indexed: 12/27/2022] Open
Abstract
The apical junction of epithelial cells can generate force to control cell geometry and perform contractile processes while maintaining barrier function and adhesion. Yet, the structural basis for force generation at the apical junction is not fully understood. Here, we describe two synaptopodin-dependent actomyosin structures that are spatially, temporally, and structurally distinct. The first structure is formed by the retrograde flow of synaptopodin initiated at the apical junction, creating a sarcomeric stress fiber that lies parallel to the apical junction. Contraction of the apical stress fiber is associated with either clustering of membrane components or shortening of junctional length. Upon junction maturation, apical stress fibers are disassembled. In mature epithelial monolayer, a motorized "contractomere" capable of "walking the junction" is formed at the junctional vertex. Actomyosin activities at the contractomere produce a compressive force evident by actin filament buckling and measurement with a new α-actinin-4 force sensor. The motility of contractomeres can adjust junctional length and change cell packing geometry during cell extrusion and intercellular movement. We propose a model of epithelial homeostasis that utilizes contractomere motility to support junction rearrangement while preserving the permeability barrier.
Collapse
Affiliation(s)
- Timothy Morris
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - Eva Sue
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - Caleb Geniesse
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - William M Brieher
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - Vivian W Tang
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| |
Collapse
|
12
|
Bachman W, Maddala R, Chakraborty A, Eldawy C, Skiba NP, Rao PV. Glucocorticoids Preferentially Influence Expression of Nucleoskeletal Actin Network and Cell Adhesive Proteins in Human Trabecular Meshwork Cells. Front Cell Dev Biol 2022; 10:886754. [PMID: 35557957 PMCID: PMC9087352 DOI: 10.3389/fcell.2022.886754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/28/2022] [Indexed: 12/02/2022] Open
Abstract
Clinical use of glucocorticoids is associated with increased intraocular pressure (IOP), a major risk factor for glaucoma. Glucocorticoids have been reported to induce changes in actin cytoskeletal organization, cell adhesion, extracellular matrix, fibrogenic activity, and mechanical properties of trabecular meshwork (TM) tissue, which plays a crucial role in aqueous humor dynamics and IOP homeostasis. However, we have a limited understanding of the molecular underpinnings regulating these myriad processes in TM cells. To understand how proteins, including cytoskeletal and cell adhesion proteins that are recognized to shuttle between the cytosolic and nuclear regions, influence gene expression and other cellular activities, we used proteomic analysis to characterize the nuclear protein fraction of dexamethasone (Dex) treated human TM cells. Treatment of human TM cells with Dex for 1, 5, or 7 days led to consistent increases (by ≥ two-fold) in the levels of various actin cytoskeletal regulatory, cell adhesive, and vesicle trafficking proteins. Increases (≥two-fold) were also observed in levels of Wnt signaling regulator (glypican-4), actin-binding chromatin modulator (BRG1) and nuclear actin filament depolymerizing protein (MICAL2; microtubule-associated monooxygenase, calponin and LIM domain containing), together with a decrease in tissue plasminogen activator. These changes were independently further confirmed by immunoblotting analysis. Interestingly, deficiency of BRG1 expression blunted the Dex-induced increases in the levels of some of these proteins in TM cells. In summary, these findings indicate that the widely recognized changes in actin cytoskeletal and cell adhesive attributes of TM cells by glucocorticoids involve actin regulated BRG1 chromatin remodeling, nuclear MICAL2, and glypican-4 regulated Wnt signaling upstream of the serum response factor/myocardin controlled transcriptional activity.
Collapse
Affiliation(s)
- William Bachman
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, United States
| | - Rupalatha Maddala
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, United States
| | - Ayon Chakraborty
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, United States
| | - Camelia Eldawy
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, United States
| | - Nikolai P. Skiba
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, United States
| | - Ponugoti V. Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, United States
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
13
|
Changes in ion channel expression and function associated with cardiac arrhythmogenic remodeling by Sorbs2. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166247. [PMID: 34487812 DOI: 10.1016/j.bbadis.2021.166247] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/03/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022]
Abstract
The Sorbin and SH3 domain-containing protein 2 (Sorbs2) is an important component of cardiomyocyte sarcomere. It has been recently reported that loss of Sorbs2 is causally associated with arrhythmogenic cardiomyopathy in human. However, the ionic mechanisms leading to cardiac arrhythmogenesis by Sorbs2 deficiency are unknown. In this study, we hypothesized that Sorbs2 plays an important role in regulating cardiac ion channel expression and function. Using electrophysiological and molecular biological approaches, we found that the Sorbs2 knockout (KO) mice progressively developed cardiac structural and electrical remodeling as early as 1 to 2 months of age and died prematurely at 5 to 7 months of age. Electrocardiographic recordings showed that Sorbs2 KO mice had conduction delays, spontaneous ventricular extrasystoles and polymorphic ventricular tachyarrhythmia. Intracellular recordings revealed abnormal action potentials with depolarized resting potential, reduced upstroke velocity, prolonged repolarization, and effective refractory period in the ventricular preparations of Sorbs2 KO mice. Patch clamp experiments demonstrated that Sorbs2 KO mice displayed distinct abnormalities in the expression and function of cardiac ion channels, including those of the voltage-gated Na+ channels, L-type Ca2+ channels, the voltage-gated K+ channels and the inward-rectifier K+ channels. Moreover, Sorbs2 physically interacted with the RNAs and/or proteins of important cardiac ion channels and directly regulated their expression in vitro. Our results indicate that Sorbs2 plays a pivotal role in the regulation of cardiac channel physiology. Loss of Sorbs2 promotes cardiac ion channelopathies and life-threatening arrhythmias.
Collapse
|
14
|
Abstract
Hepatocellular carcinoma (HCC) is a very deadly disease. HCC initiation and progression involve multiple genetic events, including the activation of proto-oncogenes and disruption of the function of specific tumor suppressor genes. Activation of oncogenes stimulates cell growth and survival, while loss-of-function mutations of tumor suppressor genes result in unrestrained cell growth. In this review, we summarize the new findings that identified novel proto-oncogenes and tumor suppressors in HCC over the past five years. These findings may inspire the development of novel therapeutic strategies to improve the outcome of HCC patients.
Collapse
|
15
|
MORC protein family-related signature within human disease and cancer. Cell Death Dis 2021; 12:1112. [PMID: 34839357 PMCID: PMC8627505 DOI: 10.1038/s41419-021-04393-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 10/06/2021] [Accepted: 11/03/2021] [Indexed: 01/03/2023]
Abstract
The microrchidia (MORC) family of proteins is a highly conserved nuclear protein superfamily, whose members contain common domain structures (GHKL-ATPase, CW-type zinc finger and coiled-coil domain) yet exhibit diverse biological functions. Despite the advancing research in previous decades, much of which focuses on their role as epigenetic regulators and in chromatin remodeling, relatively little is known about the role of MORCs in tumorigenesis and pathogenesis. MORCs were first identified as epigenetic regulators and chromatin remodelers in germ cell development. Currently, MORCs are regarded as disease genes that are involved in various human disorders and oncogenes in cancer progression and are expected to be the important biomarkers for diagnosis and treatment. A new paradigm of expanded MORC family function has raised questions regarding the regulation of MORCs and their biological role at the subcellular level. Here, we systematically review the progress of researching MORC members with respect to their domain architectures, diverse biological functions, and distribution characteristics and discuss the emerging roles of the aberrant expression or mutation of MORC family members in human disorders and cancer development. Furthermore, the illustration of related mechanisms of the MORC family has made MORCs promising targets for developing diagnostic tools and therapeutic treatments for human diseases, including cancers.
Collapse
|
16
|
Ding Y, Yang J, Chen P, Lu T, Jiao K, Tester DJ, Giudicessi JR, Jiang K, Ackerman MJ, Li Y, Wang DW, Lee H, Wang DW, Xu X. Knockout of SORBS2 Protein Disrupts the Structural Integrity of Intercalated Disc and Manifests Features of Arrhythmogenic Cardiomyopathy. J Am Heart Assoc 2020; 9:e017055. [PMID: 32808564 PMCID: PMC7660791 DOI: 10.1161/jaha.119.017055] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/13/2020] [Indexed: 12/21/2022]
Abstract
Background Sorbs2b (sorbin and SH3 domain-containing 2b) was recently identified as a cardiomyopathy gene from a zebrafish mutagenesis screen. However, cardiac functions of its mammalian ortholog remain elusive. Methods and Results We conducted a detailed expression and subcellular localization analysis of Sorbs2 ortholog in mice and a phenotypic characterization in Sorbs2 knockout mice. Sorbs2 is highly expressed in the mouse heart and encodes an adhesion junction/desmosome protein that is mainly localized to the intercalated disc. A mutation with near complete depletion of the Sorbs2 protein in mice results in phenotypes characteristic of human arrhythmogenic cardiomyopathy (ACM), including right ventricular dilation, right ventricular dysfunction, spontaneous ventricular tachycardia, and premature death. Sorbs2 is required to maintain the structural integrity of intercalated disc. Its absence resulted in profound cardiac electrical remodeling with impaired impulse conduction and action potential derangements. Targeted sequencing of human patients with ACM identified 2 rare splicing variants classified as likely pathogenic were in 2 unrelated individuals with ACM from a cohort of 59 patients with ACM. Conclusions The Sorbs2 knockout mouse manifests several key features reminiscent of human ACM. Although the candidacy of SORBS2 as a new ACM-susceptibility gene is supported by preliminary human genetics study, future validation in larger cohorts with ACM is needed.
Collapse
Affiliation(s)
- Yonghe Ding
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Jingchun Yang
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Peng Chen
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic DisordersDivision of CardiologyDepartments of Internal Medicine and Genetic Diagnosis CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Tong Lu
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Kunli Jiao
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
- Division of CardiologyXinhua HospitalShanghai Jiaotong UniversityShanghaiChina
| | | | | | - Kai Jiang
- Division of Nephrology and HypertensionMayo ClinicRochesterMN
| | - Michael J. Ackerman
- Department of Cardiovascular Medicine (Division of Heart Rhythm Services)Mayo ClinicRochesterMN
- Pediatric and Adolescent Medicine (Division of Pediatric Cardiology)Mayo ClinicRochesterMN
- Molecular Pharmacology and Experimental Therapeutics (Windland Smith Rice Sudden Death Genomics Laboratory)Mayo ClinicRochesterMN
| | - Yigang Li
- Division of CardiologyXinhua HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Dao Wu Wang
- State Key Laboratory of Reproductive MedicineClinical Center of Reproductive Medicine and Department of CardiologyFirst Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - HoN‐chi Lee
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| | - Dao Wen Wang
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic DisordersDivision of CardiologyDepartments of Internal Medicine and Genetic Diagnosis CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaolei Xu
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN
- Department of Cardiovascular MedicineMayo ClinicRochesterMN
| |
Collapse
|
17
|
Liu Y, Fan J, Yan Y, Dang X, Zhao R, Xu Y, Ding Z. JMY expression by Sertoli cells contributes to mediating spermatogenesis in mice. FEBS J 2020; 287:5478-5497. [PMID: 32279424 DOI: 10.1111/febs.15328] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 02/27/2020] [Accepted: 04/06/2020] [Indexed: 12/21/2022]
Abstract
Sertoli cells are crucial for spermatogenesis in the seminiferous epithelium because their actin cytoskeleton supports vesicular transport, cell junction formation, protein anchoring, and spermiation. Here, we show that a junction-mediating and actin-regulatory protein (JMY) affects the blood-tissue barrier (BTB) function through remodeling of the Sertoli cell junctional integrity and it also contributes to controlling endocytic vesicle trafficking. These functions are critical for the maintenance of sperm fertility since loss of Sertoli cell-specific Jmy function induced male subfertility in mice. Specifically, these mice have (a) impaired BTB integrity and spermatid adhesion in the seminiferous tubules; (b) high incidence of sperm structural deformity; and (c) reduced sperm count and poor sperm motility. Moreover, the cytoskeletal integrity was compromised along with endocytic vesicular trafficking. These effects impaired junctional protein recycling and reduced Sertoli cell BTB junctional integrity. In addition, JMY interaction with actin-binding protein candidates α-actinin1 and sorbin and SH3 domain containing protein 2 was related to JMY activity, and in turn, actin cytoskeletal organization. In summary, JMY affects the control of spermatogenesis through the regulation of actin filament organization and endocytic vesicle trafficking in Sertoli cells.
Collapse
Affiliation(s)
- Yue Liu
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, China
| | - Jiaying Fan
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, China.,Center for Experimental Medical Science Education, Shanghai Jiao Tong University School of Medicine, China
| | - Yan Yan
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, China
| | - Xuening Dang
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, China
| | - Ran Zhao
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, China
| | - Yimei Xu
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, China
| | - Zhide Ding
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
18
|
Jie R, Zhu P, Zhong J, Zhang Y, Wu H. LncRNA KCNQ1OT1 affects cell proliferation, apoptosis and fibrosis through regulating miR-18b-5p/SORBS2 axis and NF-ĸB pathway in diabetic nephropathy. Diabetol Metab Syndr 2020; 12:77. [PMID: 32905431 PMCID: PMC7469295 DOI: 10.1186/s13098-020-00585-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/21/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND It has been reported that long non-coding RNAs (lncRNAs) play vital roles in diabetic nephropathy (DN). Our study aims to research the function of lncRNA KCNQ1OT1 in DN cells and the molecular mechanism. METHODS Human glomerular mesangial cells (HGMCs) and human renal glomerular endothelial cells (HRGECs) were cultured in high glucose (30 mM) condition as models of DN cells. KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1) and miR-18b-5p levels were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The mRNA and protein levels of Sorbin and SH3 domain-containing protein 2 (SORBS2), Type IV collagen (Col-4), fibronectin (FN), transcriptional regulatory factor-beta 1 (TGF-β1), Twist, NF-κB and STAT3 were measured by qRT-PCR and western blot. Cell viability was detected by cell counting kit-8 (CCK-8) assay for selecting the proper concentration of glucose treatment. Additionally, 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) and flow cytometry assay were employed to determine cell proliferation and apoptosis, respectively. The targets of KCNQ1OT1 was predicted by online software and confirmed by dual-luciferase reporter assay. RESULTS KCNQ1OT1 and SORBS2 were elevated in DN. Both knockdown of KCNQ1OT1 and silencing of SORBS2 restrained proliferation and fibrosis and induced apoptosis in DN cells. Besides, Overexpression of SORBS2 restored the KCNQ1OT1 knockdown-mediate effects on proliferation, apoptosis and fibrosis in DN cells. In addition, miR-18b-5p served as a target of KCNQ1OT1 as well as targeted SORBS2. KCNQ1OT1 knockdown repressed NF-ĸB pathway. CONCLUSION KCNQ1OT1 regulated DN cells proliferation, apoptosis and fibrosis via KCNQ1OT1/miR-18b-5p/SORBS2 axis and NF-ĸB pathway.
Collapse
Affiliation(s)
- Ran Jie
- Department of Endocrinology, First People’s Hospital of Jingzhou, Shashi District, No. 8 Hangkong Road, Jingzhou, 434000 Hubei China
| | - Pengpeng Zhu
- Department of Anesthesiology, First People’s Hospital of Jingzhou, Jingzhou, 434000 Hubei China
| | - Jiao Zhong
- Health Management Center, First People’s Hospital of Jingzhou, Jingzhou, 434000 Hubei China
| | - Yan Zhang
- Department of Gastroenterology, First People’s Hospital of Jingzhou, Jingzhou, 434000 Hubei China
| | - Hongyan Wu
- Department of Endocrinology, First People’s Hospital of Jingzhou, Shashi District, No. 8 Hangkong Road, Jingzhou, 434000 Hubei China
| |
Collapse
|
19
|
Schlaweck AE, Tazi-Ahnini R, Ü. Basmanav FB, Mohungoo J, Pasternack-Ziach SM, Mattheisen M, Oprisoreanu AM, Humbatova A, Wolf S, Messenger A, Betz RC. Autosomal-dominant hypotrichosis with woolly hair: Novel gene locus on chromosome 4q35.1-q35.2. PLoS One 2019; 14:e0225943. [PMID: 31790498 PMCID: PMC6886801 DOI: 10.1371/journal.pone.0225943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 11/15/2019] [Indexed: 11/18/2022] Open
Abstract
Hypotrichosis simplex (HS) with and without woolly hair (WH) comprises a group of rare, monogenic disorders of hair loss. Patients present with a diffuse loss of scalp and/or body hair, which usually begins in early childhood and progresses into adulthood. Some of the patients also show hair that is tightly curled. Approximately 10 genes for autosomal recessive and autosomal dominant forms of HS have been identified in the last decade, among them five genes for the dominant form. We collected blood and buccal samples from 17 individuals of a large British family with HS and WH. After having sequenced all known dominant genes for HS in this family without the identification of any disease causing mutation, we performed a genome-wide scan, using the HumanLinkage-24 BeadChip, followed by a classical linkage analysis; and whole exome-sequencing (WES). Evidence for linkage was found for a region on chromosome 4q35.1-q35.2 with a maximum LOD score of 3.61. WES led to the identification of a mutation in the gene SORBS2, encoding sorbin and SH3 domain containing 2. Unfortunately, we could not find an additional mutation in any other patient/family with HS; and in cell culture, we could not observe any difference between cloned wildtype and mutant SORBS2 using western blotting and immunofluorescence analyses. Therefore, at present, SORBS2 cannot be considered a definite disease gene for this phenotype. However, the locus on chromosome 4q is a robust and novel finding for hypotrichosis with woolly hair. Further fine mapping and sequencing efforts are therefore warranted in order to confirm SORBS2 as a plausible HS disease gene.
Collapse
Affiliation(s)
- Annika E. Schlaweck
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Rachid Tazi-Ahnini
- Department of Infection, Immunity, and Cardiovascular Disease, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - F. Buket Ü. Basmanav
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Javed Mohungoo
- Department of Dermatology, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Sandra M. Pasternack-Ziach
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Manuel Mattheisen
- Department of Psychiatry, Psychosomatics, and Psychotherapy, University Hospital Würzburg, Würzburg, Germany
| | - Ana-Maria Oprisoreanu
- Department of Neuropathology and Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Aytaj Humbatova
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Sabrina Wolf
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Andrew Messenger
- Department of Dermatology, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | - Regina C. Betz
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
- * E-mail:
| |
Collapse
|
20
|
Han L, Huang C, Zhang S. The RNA-binding protein SORBS2 suppresses hepatocellular carcinoma tumourigenesis and metastasis by stabilizing RORA mRNA. Liver Int 2019; 39:2190-2203. [PMID: 31365778 DOI: 10.1111/liv.14202] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/13/2019] [Accepted: 07/25/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Numerous studies have revealed that dysregulation of RNA-binding protein (RBP) expression is causally linked with human cancer tumourigenesis. However, the detailed biological effect and underlying mechanisms of most RBPs remain unclear. METHODS Expression of sorbin and SH3 domain-containing 2 (SORBS2) in hepatocellular carcinoma (HCC) was detected by qRT-PCR, immunohistochemistry assay and Western blot assay. Proliferation, migration, invasion and cell cycle progression of HCC cells were measured by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, colony-forming assay, Transwell assay and flow cytometry assay respectively. A xenograft model and metastatic model were established to evaluate the proliferation and metastasis of HCC cells in vivo. Western blot assays were performed to assess the expression of epithelial-mesenchymal transition markers. Luciferase reporter assay, RNA immunoprecipitation and pull-down assay elucidated the effect of SORBS2 on one of its downstream genes. RESULTS The expression of SORBS2 was significantly decreased in HCC and was associated with metastasis, advanced TNM clinical stage and poor clinical outcome of HCC patients. Furthermore, our results suggested that SORBS2 inhibited HCC cell proliferation, invasion, migration and EMT both in vivo and in vitro. Mechanistically, we revealed that retinoic acid receptor-related orphan receptor (RORA) was a major target of SORBS2 and was critical to sustaining the antitumour effect of SORBS2 on HCC cells. SORBS2 reduced RORA mRNA degradation by directly binding to the 3'UTR of RORA mRNA. CONCLUSIONS In this study, we found for the first time that SORBS2 contributed to the suppression of HCC tumourigenesis and metastasis via post-transcriptional regulation of RORA expression as an RBP.
Collapse
Affiliation(s)
- Lili Han
- Department of Oncology, College of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Chen Huang
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, China
| | - Shuqun Zhang
- Department of Oncology, College of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
21
|
Fu K, Nakano H, Morselli M, Chen T, Pappoe H, Nakano A, Pellegrini M. A temporal transcriptome and methylome in human embryonic stem cell-derived cardiomyocytes identifies novel regulators of early cardiac development. Epigenetics 2018; 13:1013-1026. [PMID: 30240284 PMCID: PMC6342070 DOI: 10.1080/15592294.2018.1526029] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/30/2018] [Accepted: 09/09/2018] [Indexed: 02/07/2023] Open
Abstract
Stem cell-based cardiogenesis has become a powerful tool to enhance our understanding of cardiac development and test novel therapeutics for cardiovascular diseases. However, transcriptional and epigenetic regulation of multiple transitional stages from pluripotent cells to committed cardiomyocytes has not yet been fully characterized. To characterize how transcription factors, lincRNAs and DNA methylation change at temporal developmental stages, and identify potential novel regulators during cardiogenesis. We utilized a previously reported protocol that yields human cardiomyocytes (hCM) with more than 90% purity from human Embryonic Stem Cells (hESC). Leveraging the purity of cells resulting from this protocol, we systematically examined how gene expression and DNA methylation programs change at temporal developmental stages during cardiogenesis. Our results provide a comprehensive view of expression changes during cardiogenesis that extend previous studies, allowing us to identify key transcription factors as well as lincRNAs that are strongly associated with cardiac differentiation. Moreover, we incorporated a simple but powerful method to screen for novel regulators of cardiogenesis solely based on expression changes and found four novel cardiac-related transcription factors, i.e., SORBS2, MITF, DPF3, and ZNF436, which have no or few prior literature reports and we were able to validate using siRNA. Our strategy of identifying novel regulators of cardiogenesis can also be easily implemented in other stem cell-based systems. Our results provide a valuable resource for understanding cardiogenesis that extends previous findings by leveraging the purity of our cell lines, which allowed us to identify four novel cardiac-related regulators.
Collapse
Affiliation(s)
- Kai Fu
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Haruko Nakano
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
| | - Marco Morselli
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Tiffany Chen
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Herman Pappoe
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Atsuschi Nakano
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Cardiology Division, School of Medicine, University of California, Los Angeles, CA, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA, USA
| |
Collapse
|
22
|
Kuroda M, Ueda K, Kioka N. Vinexin family (SORBS) proteins regulate mechanotransduction in mesenchymal stem cells. Sci Rep 2018; 8:11581. [PMID: 30068914 PMCID: PMC6070524 DOI: 10.1038/s41598-018-29700-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 07/17/2018] [Indexed: 01/09/2023] Open
Abstract
The stiffness of extracellular matrix (ECM) directs the differentiation of mesenchymal stem cells (MSCs) through the transcriptional co-activators Yes-associated protein (YAP) and transcriptional coactivator with a PDZ-binding motif (TAZ). Although a recent study revealed the involvement of vinexin α and CAP (c-Cbl-associated proteins), two of vinexin (SORBS) family proteins that bind to vinculin, in mechanosensing, it is still unclear whether these proteins regulate mechanotransduction and differentiation of MSCs. In the present study, we show that both vinexin α and CAP are necessary for the association of vinculin with the cytoskeleton and the promotion of YAP/TAZ nuclear localization in MSCs grown on rigid substrates. Furthermore, CAP is involved in the MSC differentiation in a stiffness-dependent manner, whereas vinexin depletion suppresses adipocyte differentiation independently of YAP/TAZ. These observations reveal a critical role of vinexin α and CAP in mechanotransduction and MSC differentiation.
Collapse
Affiliation(s)
- Mito Kuroda
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto, 606-8502, Japan
| | - Kazumitsu Ueda
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto, 606-8502, Japan
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Sakyo, Kyoto, 606-8507, Japan
| | - Noriyuki Kioka
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto, 606-8502, Japan.
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Sakyo, Kyoto, 606-8507, Japan.
| |
Collapse
|
23
|
Pan Z, Ding Q, Guo Q, Guo Y, Wu L, Wu L, Tang M, Yu H, Zhou F. MORC2, a novel oncogene, is upregulated in liver cancer and contributes to proliferation, metastasis and chemoresistance. Int J Oncol 2018; 53:59-72. [PMID: 29620211 PMCID: PMC5958890 DOI: 10.3892/ijo.2018.4333] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/22/2018] [Indexed: 12/20/2022] Open
Abstract
Microrchidia 2 (MORC2) is important in DNA damage repair and lipogenesis, however, the clinical and functional role of MORC2 in liver cancer remains to be fully elucidated. The aim the present study was to clarify the role of MORC2 in liver cancer. Expression profile analysis, immunohistochemical staining, reverse transcription-quantitative polymerase chain reaction analysis and western blot analysis were performed to evaluate the levels of MORC2 in liver cancer patient specimens and cell lines; subsequently the expression of MORC2 was suppressed or increased in liver cancer cells and the effects of MORC2 on the cancerous transformation of liver cancer cells were examined in vitro and in vivo. MORC2 was upregulated in liver cancer tissues, and the upregulation was associated with certain clinicopathologic features of patients with liver cancer. MORC2 knockdown caused marked inhibition of liver cancer cell proliferation and clonogenicity, whereas the overexpression of MORC2 substantially promoted liver cancer cell proliferation. In addition, the knockdown of MORC2 inhibited the migratory and invasive ability of liver cancer cells, whereas increased migration and invasion rates were observed in cells with ectopic expression of MORC2. In a model of nude mice, the overexpression of MORC2 promoted tumorigenicity and markedly enhanced pulmonary metastasis of liver cancer. Furthermore, MORC2 regulated apoptosis and its expression level had an effect on the sensitivity of liver cancer cells to doxorubicin, 5-fluorouracil and cisplatin. Mechanically, MORC2 modulated the mitochondrial apoptotic pathway, possibly in a p53-dependent manner, and its dysregulation also resulted in the abnormal activation of the Hippo pathway. For the first time, to the best of our knowledge, the present study confirmed that MORC2 was a novel oncogene in liver cancer. These results provide useful insight into the mechanism underlying the tumorigenesis and progression of liver cancer, and offers clues into potential novel liver cancer therapies.
Collapse
Affiliation(s)
- Zhihong Pan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060
| | - Qianshan Ding
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060
| | - Qian Guo
- Hepatic Disease Institute, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430061
| | - Yong Guo
- College of Biotechnology, Guilin Medical University, Guilin, Guanxi 541004
| | - Lianlian Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060
| | - Lu Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060
| | - Meng Tang
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Clinical Cancer Study Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Honggang Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060
| | - Fuxiang Zhou
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Clinical Cancer Study Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
24
|
Tong Y, Li Y, Gu H, Wang C, Liu F, Shao Y, Li F. HSF1, in association with MORC2, downregulates ArgBP2 via the PRC2 family in gastric cancer cells. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1104-1114. [PMID: 29339121 DOI: 10.1016/j.bbadis.2018.01.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 01/07/2018] [Accepted: 01/08/2018] [Indexed: 11/30/2022]
Abstract
Arg Kinase-binding protein 2 (ArgBP2) is considered to be a scaffold protein that coordinates multiple signaling pathways converging on cell adhesion and actin cytoskeletal organization. It also plays an important role in blocking cancer metastasis as a potential tumor suppressor. However, its regulation mechanisms in tumor migration, especially in gastric cancer, are not fully understood. Here, we identified an ArgBP2 enhancer and showed that heat shock factor 1 (HSF1) directly interacted with microrchidia CW-type zinc finger 2 (MORC2) and bound to the enhancer of ArgBP2. HSF1 was found to promote proliferation, migration and invasion of gastric cancer cells. HSF1 or/and MORC2 increased recruitment of the polycomb repressive complex 2 (PRC2), particularly enhancer of zeste homolog 2 (EZH2), to the ArgBP2 enhancer and catalyzed tri-methylation of lysine 27 on histone H3 (H3K27me3), leading to transcriptional repression of ArgBP2. In addition, HSF1 and MORC2-induced migration and invasion in gastric cancer cells was dependent on ArgBP2 or EZH2. Clinical data exhibited a negative correlation of ArgBP2 with MORC2, HSF1, and EZH2. Our results thus contribute to the knowledge of the regulatory mechanism of HSF1 in down-regulating ArgBP2, providing new insight into the HSF1&MORC2-PRC2-ArgBP2 signaling pathway and a better understanding of their functions in gastric cancer cells.
Collapse
Affiliation(s)
- Yuxin Tong
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China; Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China.
| | - Yan Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Hui Gu
- Department of Key Laboratory of Health Ministry for Congenital Malformation Shengjing Hospital, China Medical University, Shenyang, Liaoning 110004, China
| | - Chunyu Wang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Funan Liu
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yangguang Shao
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
25
|
Zhao L, Wang W, Huang S, Yang Z, Xu L, Yang Q, Zhou X, Wang J, Shen Q, Wang C, Le X, Feng M, Zhou N, Lau WB, Lau B, Yao S, Yi T, Wang X, Zhao X, Wei Y, Zhou S. The RNA binding protein SORBS2 suppresses metastatic colonization of ovarian cancer by stabilizing tumor-suppressive immunomodulatory transcripts. Genome Biol 2018; 19:35. [PMID: 29548303 PMCID: PMC5857099 DOI: 10.1186/s13059-018-1412-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/22/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Ovarian cancer constitutes one of the most lethal gynecologic malignancies for females. Currently, early detection strategies and therapeutic options for ovarian cancer are far from satisfactory, leading to high diagnosis rates at late stages and disease relapses. New avenues of therapy are needed that target key processes in ovarian cancer progression. While a variety of non-coding RNAs have been proven to regulate ovarian cancer metastatic progression, the functional roles of RNA-binding proteins (RBPs) in this process are less well defined. RESULTS In this study, we identify that the RBP sorbin and SH3 domain containing 2 (SORBS2) is a potent suppressor of ovarian cancer metastatic colonization. Mechanistic studies show that SORBS2 binds the 3' untranslated regions (UTRs) of WFDC1 (WAP four-disulfide core domain 1) and IL-17D (Interleukin-17D), two secreted molecules that are shown to act as metastasis suppressors. Enhanced expression of either WFDC1 or IL-17D potently represses SORBS2 depletion-mediated cancer metastasis promotion. By enhancing the stability of these gene transcripts, SORBS2 suppresses ovarian cancer invasiveness and affects monocyte to myeloid-derived suppressor cell and M2-like macrophage polarization, eliciting a tumor-suppressive immune microenvironment. CONCLUSIONS Our data illustrate a novel post-transcriptional network that links cancer progression and immunomodulation within the tumor microenvironment through SORBS2-mediated transcript stabilization.
Collapse
Affiliation(s)
- Linjie Zhao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, People's Republic of China
| | - Wei Wang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Shuang Huang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, People's Republic of China
| | - Zhengnan Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, People's Republic of China
| | - Lian Xu
- Department of Pathology, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qilian Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, People's Republic of China
| | - Xiu Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, People's Republic of China
| | - Jinjin Wang
- College of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Qiuhong Shen
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, People's Republic of China
| | - Chenlu Wang
- College of Life Sciences, Sichuan University, Chengdu, People's Republic of China
| | - Xiaobing Le
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, People's Republic of China
| | - Min Feng
- Department of Pathology, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Nianxin Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, People's Republic of China
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Bonnie Lau
- Department of Surgery, Emergency Medicine, Kaiser Santa Clara Medical Center, Affiliate of Stanford University, Santa Clara, CA, USA
| | - Shaohua Yao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, People's Republic of China
| | - Tao Yi
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, People's Republic of China
| | - Xin Wang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Xia Zhao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, People's Republic of China
| | - Yuquan Wei
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, People's Republic of China
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
26
|
Loveless T, Qadota H, Benian GM, Hardin J. Caenorhabditis elegans SORB-1 localizes to integrin adhesion sites and is required for organization of sarcomeres and mitochondria in myocytes. Mol Biol Cell 2017; 28:3621-3633. [PMID: 28978740 PMCID: PMC5706990 DOI: 10.1091/mbc.e16-06-0455] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 09/18/2017] [Accepted: 09/25/2017] [Indexed: 01/07/2023] Open
Abstract
We have identified and characterized sorb-1, the only sorbin and SH3 domain-containing protein family member in Caenorhabditis elegans SORB-1 is strongly localized to integrin adhesion complexes in larvae and adults, including adhesion plaques and dense bodies (Z-disks) of striated muscles and attachment plaques of smooth muscles. SORB-1 is recruited to the actin-binding, membrane-distal regions of dense bodies via its C-terminal SH3 domains in an ATN-1(α-actinin)- and ALP-1(ALP/Enigma)-dependent manner, where it contributes to the organization of sarcomeres. SORB-1 is also found in other tissues known to be under mechanical stress, including stress fibers in migratory distal tip cells and the proximal gonad sheath, where it becomes enriched in response to tissue distention. We provide evidence for a novel role for sorbin family proteins: SORB-1 is required for normal positioning of the mitochondrial network in muscle cells. Finally, we demonstrate that SORB-1 interacts directly with two other dense body components, DEB-1(vinculin) and ZYX-1(zyxin). This work establishes SORB-1 as a bona fide sorbin family protein-one of the late additions to the dense body complex and a conserved regulator of body wall muscle sarcomere organization and organelle positioning.
Collapse
Affiliation(s)
- Timothy Loveless
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
| | - Hiroshi Qadota
- Department of Pathology, Emory University, Atlanta, GA 30322
| | - Guy M Benian
- Department of Pathology, Emory University, Atlanta, GA 30322
| | - Jeff Hardin
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
27
|
Fredriksson-Lidman K, Van Itallie CM, Tietgens AJ, Anderson JM. Sorbin and SH3 domain-containing protein 2 (SORBS2) is a component of the acto-myosin ring at the apical junctional complex in epithelial cells. PLoS One 2017; 12:e0185448. [PMID: 28961272 PMCID: PMC5621683 DOI: 10.1371/journal.pone.0185448] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/12/2017] [Indexed: 12/30/2022] Open
Abstract
SORBS2 is a scaffolding protein associated with Abl/Arg non-receptor tyrosine kinase pathways and is known to interact with actin and several other cytoskeletal proteins in various cell types. Previous BioID proximity labeling of tight and adherens junction proteins suggested that SORBS2 is a component of the apical junction complex of epithelial cells. We asked whether SORBS2 plays a previously unappreciated role in controlling perijunctional actin and tight junction barrier function. Using super resolution imaging we confirmed that SORBS2 is localized at the apical junction complex but farther from the membrane than ZO-1 and located partially overlapping both the tight- and adherens junctions with a periodic concentration that alternates with myosin IIB in polarized epithelial cells. Overexpression of GFP-SORBS2 recruited alpha-actinin, vinculin and N-WASP, and possibly CIP4 to cellular junctions. However, CRISPR-Cas9 knock-out of SORBS2 did not alter the localization- or immunofluorescent staining intensity of these or several other junctional- and cytoskeletal proteins. SORBS2 knock-out also did not affect the barrier function as measured by TER and dextran flux; nor did it change actin-dependent junction re-assembly as measured by Ca2+-switch and Latrunculin-B wash-out assays. The kinetics of HGF-induced cell scattering and wound healing, and dextran flux increase induced by PDGF also were unaffected by SORBS2 knock-out. SORBS2 concentrates with apical junctional actin that accumulates in response to knock-down of ZO-1 and ZO-2. In spite of our finding that SORBS2 is clearly a component of the apical junction complex, it does not appear to be required for either normal tight- or adherens junction assembly, structure or function or for growth factor-mediated changes in tight junction dynamics.
Collapse
Affiliation(s)
- Karin Fredriksson-Lidman
- Laboratory of Tight Junction Structure and Function, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| | - Christina M. Van Itallie
- Laboratory of Tight Junction Structure and Function, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Amber J. Tietgens
- Laboratory of Tight Junction Structure and Function, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - James M. Anderson
- Laboratory of Tight Junction Structure and Function, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
28
|
Ichikawa T, Kita M, Matsui TS, Nagasato AI, Araki T, Chiang SH, Sezaki T, Kimura Y, Ueda K, Deguchi S, Saltiel AR, Kioka N. Vinexin family (SORBS) proteins play different roles in stiffness-sensing and contractile force generation. J Cell Sci 2017; 130:3517-3531. [PMID: 28864765 DOI: 10.1242/jcs.200691] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 08/10/2017] [Indexed: 12/17/2022] Open
Abstract
Vinexin, c-Cbl associated protein (CAP) and Arg-binding protein 2 (ArgBP2) constitute an adaptor protein family called the vinexin (SORBS) family that is targeted to focal adhesions (FAs). Although numerous studies have focused on each of the SORBS proteins and partially elucidated their involvement in mechanotransduction, a comparative analysis of their function has not been well addressed. Here, we established mouse embryonic fibroblasts that individually expressed SORBS proteins and analysed their functions in an identical cell context. Both vinexin-α and CAP co-localized with vinculin at FAs and promoted the appearance of vinculin-rich FAs, whereas ArgBP2 co-localized with α-actinin at the proximal end of FAs and punctate structures on actin stress fibers (SFs), and induced paxillin-rich FAs. Furthermore, both vinexin-α and CAP contributed to extracellular matrix stiffness-dependent vinculin behaviors, while ArgBP2 stabilized α-actinin on SFs and enhanced intracellular contractile forces. These results demonstrate the differential roles of SORBS proteins in mechanotransduction.
Collapse
Affiliation(s)
- Takafumi Ichikawa
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan.,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Sakyo, Kyoto 606-8507, Japan
| | - Masahiro Kita
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Tsubasa S Matsui
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Showa, Nagoya 466-8555, Japan.,Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | - Ayaka Ichikawa Nagasato
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Tomohiko Araki
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Showa, Nagoya 466-8555, Japan
| | - Shian-Huey Chiang
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Takuhito Sezaki
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Yasuhisa Kimura
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Kazumitsu Ueda
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan.,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Sakyo, Kyoto 606-8507, Japan
| | - Shinji Deguchi
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Showa, Nagoya 466-8555, Japan.,Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | - Alan R Saltiel
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Noriyuki Kioka
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan .,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Sakyo, Kyoto 606-8507, Japan
| |
Collapse
|
29
|
Braun GS, Kuszka A, Dau C, Kriz W, Moeller MJ. Interaction of atypical cadherin Fat1 with SoHo adaptor proteins CAP/ponsin and ArgBP2. Biochem Biophys Res Commun 2016; 472:88-94. [PMID: 26903299 DOI: 10.1016/j.bbrc.2016.02.069] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 02/17/2016] [Indexed: 12/11/2022]
Abstract
Mammalian Fat1 is a giant atypical cadherin/tumor suppressor involved in the regulation of cellular orientation, migration, and growth. Fat1 is implicated in the development of the brain, eye, and kidney. Altered expression or mutations of FAT1 are also associated with cancer and facioscapulohumeral muscular dystrophy (FSHD). Yet, the mechanistic functions of this pathway remain incompletely understood. Here, we report the identification of Sorbin-homology (SoHo) proteins as novel interaction partners of Fat1 by virtue of a yeast-two-hybrid screen. SoHo proteins play diverse roles as adaptor proteins in cell signaling, cell adhesion and sarcomere architecture, including altered expression in cancer and FSHD. Specifically, we found SoHo proteins CAP/ponsin-1 and -2 (Sorbs1) and ArgBP2 (Sorbs2) to interact with the cytoplasmic domain of Fat1. We mapped the interaction to a prolin-rich classic type II PXXP motif within Fat1 and to the three Src-homology (SH3) domains within SoHo proteins using mutant expression in yeast, pulldown assays, and cell culture. Functionally, endogenous ponsin-2 expression of NRK-52E cells at cellular leading edges was lost upon knockdown of Fat1. In summary, our data point to an interaction of Fat1 with SoHo proteins that is able to recruit SoHo proteins to sites of Fat1 expression.
Collapse
Affiliation(s)
- Gerald S Braun
- Division of Nephrology and Immunology, RWTH Aachen University, Germany.
| | | | - Cécile Dau
- Kaiser-Franz-Josef-Spital mit Gottfried von Preyer'schem Kinderspital, Vienna, Austria
| | - Wilhelm Kriz
- Institute for Neuroanatomy, Medical Faculty Mannheim of the University of Heidelberg, Germany
| | - Marcus J Moeller
- Division of Nephrology and Immunology, RWTH Aachen University, Germany
| |
Collapse
|
30
|
Zhang Q, Gao X, Li C, Feliciano C, Wang D, Zhou D, Mei Y, Monteiro P, Anand M, Itohara S, Dong X, Fu Z, Feng G. Impaired Dendritic Development and Memory in Sorbs2 Knock-Out Mice. J Neurosci 2016; 36:2247-60. [PMID: 26888934 PMCID: PMC4756157 DOI: 10.1523/jneurosci.2528-15.2016] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 12/21/2015] [Accepted: 01/13/2016] [Indexed: 12/27/2022] Open
Abstract
Intellectual disability is a common neurodevelopmental disorder characterized by impaired intellectual and adaptive functioning. Both environmental insults and genetic defects contribute to the etiology of intellectual disability. Copy number variations of SORBS2 have been linked to intellectual disability. However, the neurobiological function of SORBS2 in the brain is unknown. The SORBS2 gene encodes ArgBP2 (Arg/c-Abl kinase binding protein 2) protein in non-neuronal tissues and is alternatively spliced in the brain to encode nArgBP2 protein. We found nArgBP2 colocalized with F-actin at dendritic spines and growth cones in cultured hippocampal neurons. In the mouse brain, nArgBP2 was highly expressed in the cortex, amygdala, and hippocampus, and enriched in the outer one-third of the molecular layer in dentate gyrus. Genetic deletion of Sorbs2 in mice led to reduced dendritic complexity and decreased frequency of AMPAR-miniature spontaneous EPSCs in dentate gyrus granule cells. Behavioral characterization revealed that Sorbs2 deletion led to a reduced acoustic startle response, and defective long-term object recognition memory and contextual fear memory. Together, our findings demonstrate, for the first time, an important role for nArgBP2 in neuronal dendritic development and excitatory synaptic transmission, which may thus inform exploration of neurobiological basis of SORBS2 deficiency in intellectual disability. SIGNIFICANCE STATEMENT Copy number variations of the SORBS2 gene are linked to intellectual disability, but the neurobiological mechanisms are unknown. We found that nArgBP2, the only neuronal isoform encoded by SORBS2, colocalizes with F-actin at neuronal dendritic growth cones and spines. nArgBP2 is highly expressed in the cortex, amygdala, and dentate gyrus in the mouse brain. Genetic deletion of Sorbs2 in mice leads to impaired dendritic complexity and reduced excitatory synaptic transmission in dentate gyrus granule cells, accompanied by behavioral deficits in acoustic startle response and long-term memory. This is the first study of Sorbs2 function in the brain, and our findings may facilitate the study of neurobiological mechanisms underlying SORBS2 deficiency in the development of intellectual disability.
Collapse
Affiliation(s)
- Qiangge Zhang
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142
| | - Xian Gao
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Key Laboratory of Brain Functional Genomics (Ministry of Education and Science and Technology Commission of Shanghai Municipality), Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142
| | - Chenchen Li
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142
| | - Catia Feliciano
- Champalimaud Neuroscience Programme, Champalimaud Center for the Unknown, Lisbon 1400-038, Portugal
| | - Dongqing Wang
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Dingxi Zhou
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, School of Life Sciences, Peking University, Beijing 100871, China, and
| | - Yuan Mei
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Patricia Monteiro
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142
| | - Michelle Anand
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Shigeyoshi Itohara
- Laboratory of Behavioral Genetics, RIKEN Brain Science Institute, Wako 351-0198, Japan
| | - Xiaowei Dong
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Science and Technology Commission of Shanghai Municipality), Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China
| | - Zhanyan Fu
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, Key Laboratory of Brain Functional Genomics (Ministry of Education and Science and Technology Commission of Shanghai Municipality), Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China, Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts 02142,
| |
Collapse
|
31
|
Chi Y, Huang S, Liu M, Guo L, Shen X, Wu J. Cyclin D3 predicts disease-free survival in breast cancer. Cancer Cell Int 2015; 15:89. [PMID: 26412984 PMCID: PMC4583737 DOI: 10.1186/s12935-015-0245-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 09/17/2015] [Indexed: 01/27/2023] Open
Abstract
Background Cyclin D3, which induces progression through the G1 phase of the cell cycle, is a regulator of Cyclin-dependent kinases 4 and 6. Previous studies revealed that abnormal expression of Cyclin D3 was found in many different cancers. However, the role of Cyclin D3 in breast cancer (BC) remains unknown. The aim of this study is to examine the expression pattern of Cyclin D3 in BC and to evaluate its biological role and clinical significance in prognosis prediction. The mechanism involved is also evaluated. Methods Immunohistochemical staining was used to detect the expression of Cyclin D3. qRT-PCR was used to detect the mRNA level of Cyclin D3 in BC tissues and BC cell lines. Transwell assay was used to examine the role of Cyclin D3 in the migration and invasion of BC cells. Mass Spectrometry was used to search for the interacting protein with Cyclin D3. Co-Immunoprecipitation assay and GST-Pull Down assay were used to validate the interaction of Cyclin D3 and its interaction protein. Results Through detecting Cyclin D3 expression in 243 breast cancer patients’ tissue array, we found Cyclin D3 expression was correlated with ER status (p = 0.000), PR status (p = 0.001), HER2 status (p = 0.002) and tumor differentiation (p = 0.045). The Kaplan–Meier survival curves indicated that the disease free survival (DFS) was significantly poor in high Cyclin D3 expression BC patients (p = 0.004). Furthermore, expression of Cyclin D3 was significantly associated with BC prognosis and was shown to be an independent prognostic marker in breast cancer (p = 0.028). By IHC staining and qPCR detection, Cyclin D3 expression was found to be down-regulated both in BC tissues and in BC cell lines compared with the corresponding normal controls. Further investigation showed Cyclin D3 was involved in the metastasis of BC cells and physically interacted with actin in vivo and in vitro. Conclusion Our studies revealed that Cyclin D3 was upregulated in breast cancer and represented a novel predictor of BC prognosis. Electronic supplementary material The online version of this article (doi:10.1186/s12935-015-0245-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yayun Chi
- Department of Breast Surgery, Breast Cancer Institute, Fudan University Shanghai Cancer Center, Building 7, No. 270 Dong An Road, Shanghai, 200032 China
| | - Sheng Huang
- Department of Breast Surgery, Breast Cancer Institute, Fudan University Shanghai Cancer Center, Building 7, No. 270 Dong An Road, Shanghai, 200032 China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Mengying Liu
- Department of Breast Surgery, Breast Cancer Institute, Fudan University Shanghai Cancer Center, Building 7, No. 270 Dong An Road, Shanghai, 200032 China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Liang Guo
- Department of Breast Surgery, Breast Cancer Institute, Fudan University Shanghai Cancer Center, Building 7, No. 270 Dong An Road, Shanghai, 200032 China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Xuxia Shen
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
| | - Jiong Wu
- Department of Breast Surgery, Breast Cancer Institute, Fudan University Shanghai Cancer Center, Building 7, No. 270 Dong An Road, Shanghai, 200032 China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| |
Collapse
|