Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Reprod Biol Endocrinol ; 19(1): 48, 2021 Mar 23.
Article in English | MEDLINE | ID: mdl-33757527

ABSTRACT

BACKGROUND: Proper differentiation of trophoblasts in the human placenta is essential for a successful pregnancy, whereas abnormal regulation of this process may lead to adverse pregnancy outcomes, especially preeclampsia (PE). However, the underlying mechanism of trophoblast differentiation remains unclear. Previous studies have reported the involvement of alpha-actinin-4 (ACTN4) in the actin cytoskeleton dynamics and motility. Hence, we hypothesized that ACTN4 may act as an important regulator in the normal proliferation and differentiation of trophoblasts during early pregnancy. METHOD: To test this hypothesis, we collected villous tissues from women undergoing a legal pregnancy termination during 6-10 weeks of gestation and explanted them for cell culture and siRNA transfection. We also obtained placental tissues from PE patients and healthy pregnant women and isolated the primary cytotrophoblast (CTB) cells. The expression of ACTN4 in the CTBs of placental villi and during the differentiation of CTBs into STBs was detected by immunofluorescence, immunohistochemistry (IHC), and EdU proliferation assays. Besides, villous explant, Matrigel invasion, transwell migration assay, and Wound-healing assay were performed to identify the possible role of ACTN4 in the outgrowth of explants and the invasion, migration, and proliferation of cell column trophoblasts (CCTs). Western blot analysis was carried out to compare the protein expression level of AKT, Snail activities, and epithelial-to-mesenchymal transition (EMT) in the villi or HTR8/SVneo cells with ACTN4 knockdown. RESULTS: ACTN4 was highly expressed in CTB cells and interstitial extravillous trophoblast (iEVT) cells but not found in the syncytiotrophoblast (STB) cells in the first trimester villi. Downregulation of ACTN4 led to reduced trophoblast proliferation and explant outgrowth ex vivo, as well as iEVT invasion and migration in vitro due to disrupt of actin filaments organization. Such ACTN4 inhibition also decreased AKT and Snail activities and further impeded the EMT process. In addition, ACTN4 expression was found to be downregulated in the iEVTs from preeclamptic placentas. CONCLUSIONS: Our findings suggest that ACTN4 may act as an important regulator of trophoblast proliferation and differentiation during early pregnancy, and dysregulation of this protein may contribute to preeclampsia pathogenesis.


Subject(s)
Actinin/biosynthesis , Cell Differentiation/physiology , Cell Proliferation/physiology , Chorionic Villi/metabolism , Trophoblasts/metabolism , Actinin/antagonists & inhibitors , Actinin/genetics , Adolescent , Adult , Cell Line, Transformed , Cells, Cultured , Female , Gene Knockdown Techniques/methods , Humans , Pregnancy , Young Adult
2.
J Exp Clin Cancer Res ; 36(1): 172, 2017 Dec 02.
Article in English | MEDLINE | ID: mdl-29197410

ABSTRACT

BACKGROUND: Pharmacology-based target identification has become a novel strategy leading to the discovery of novel pathological biomarkers. Ellagic acid (EA), a dietary polyphenol compound, exhibits potent anticancer activities; however, the underlying mechanisms remain unclear. The current study sought to determine the role and regulation of ACTN4 expression in human breast cancer metastasis and EA-based therapy. METHODS: The anti-metastasis ability of EA was validated by MMTV-PyMT mice and in vitro cell models. Drug affinity responsive target stability (DARTS) was utilized to identify ACTN4 as the direct target of EA. The metastatic regulated function of ACTN4 were assessed by cancer stem cells (CSCs)-related assays, including mammosphere formation, tumorigenic ability, reattachment differentiation, and signaling pathway analysis. The mechanisms of ACTN4 on ß-catenin stabilization were investigated by western blotting, co-immunoprecipitation and ubiquitination assays. The clinical significance of ACTN4 was based on human tissue microarray (TMA) analysis and The Cancer Genome Atlas (TCGA) database exploration. RESULTS: EA inhibited breast cancer growth and metastasis via directly targeting ACTN4 in vitro and in vivo, and was accompanied by a limited CSC population. ACTN4 knockdown resulted in the blockage of malignant cell proliferation, colony formation, and ameliorated metastasis potency. ACTN4-positive CSCs exhibited a higher ESA+ proportion, increased mammosphere-formation ability, and enhanced in vivo tumorigenesis ability. Mechanism exploration revealed that interruption of ACTN4/ß-catenin interaction will result in the activation of ß-catenin proteasome degradation. Increased ACTN4 expression was directly associated with the advanced cancer stage, an increased incidence of metastasis, and poor overall survival period. CONCLUSIONS: Taken together, our results suggest that ACTN4 plays an important role in breast CSCs-related metastasis and is a novel therapeutic target of EA treatment.


Subject(s)
Actinin/antagonists & inhibitors , Breast Neoplasms/drug therapy , Ellagic Acid/pharmacology , Neoplastic Stem Cells/drug effects , beta Catenin/metabolism , Actinin/biosynthesis , Actinin/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , MCF-7 Cells , Mice , Mice, Transgenic , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Random Allocation , Transfection , Xenograft Model Antitumor Assays
3.
Mol Biosyst ; 13(8): 1469-1480, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28604846

ABSTRACT

Traditional Chinese medicine (TCM) preparations have significant effects on some refractory diseases; however, these compositions are complex and their mechanisms are unknown. Identification of the active components in these preparations is essential. The mortality rate for heart failure (HF) has been increasing in recent years, and myocardial dysfunction (MD) has been proved to be the pathological basis of HF. Yixinshu Capsule (YXSC) is a multi-component oral drug with therapeutic effects on HF. However, the key active components are still unclear. In this study, YXSC intestinal absorption liquid (IAL) was used and 62 compounds were identified by an analytical chemistry approach. Then, a compound - target - function network was established with a bioinformatics analysis tool. Finally, a cell model of MD on human-induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) was used to verify the therapeutic effects of the active components of YXSC. Schisandrin A (Sch A) and schisandrin B (Sch B) were demonstrated to be the active components of YXSC by attenuating endothelin-1 (ET-1)-induced contraction dysfunction, brain natriuretic peptide (BNP) content elevation, and the morphological changes of hiPS-CMs. For the first time, our data illustrate the potent protective effects of Sch A and Sch B on ET-1-induced dysfunctional hiPS-CMs and revealed their effective targets and pathways. The integrative approach used in our study was applied to identify active components in TCM preparations and excavate the possible mechanisms.


Subject(s)
Cardiotonic Agents/pharmacology , Cyclooctanes/pharmacology , Drugs, Chinese Herbal/chemistry , Endothelin Receptor Antagonists/pharmacology , Lignans/pharmacology , Myocytes, Cardiac/drug effects , Polycyclic Compounds/pharmacology , Actinin/antagonists & inhibitors , Actinin/genetics , Actinin/metabolism , Animals , Bosentan , Cardiotonic Agents/chemistry , Cardiotonic Agents/isolation & purification , Cell Differentiation , Cell Line , Cyclooctanes/chemistry , Cyclooctanes/isolation & purification , Drugs, Chinese Herbal/pharmacology , Endothelin Receptor Antagonists/chemistry , Endothelin Receptor Antagonists/isolation & purification , Endothelin-1/antagonists & inhibitors , Endothelin-1/pharmacology , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Intestinal Mucosa/metabolism , Intestines/drug effects , Lignans/chemistry , Lignans/isolation & purification , Male , Medicine, Chinese Traditional , Metabolic Networks and Pathways/drug effects , Metabolome , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Natriuretic Peptide, Brain/antagonists & inhibitors , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Polycyclic Compounds/chemistry , Polycyclic Compounds/isolation & purification , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacology , Troponin T/antagonists & inhibitors , Troponin T/genetics , Troponin T/metabolism
4.
PLoS One ; 10(4): e0120616, 2015.
Article in English | MEDLINE | ID: mdl-25860875

ABSTRACT

α-Actinins (ACTNs) are known to crosslink actin filaments at focal adhesions in migrating cells. Among the four isoforms of mammalian ACTNs, ACTN1 and ACTN4 are ubiquitously expressed. Recently, ACTN4 was reported to enhance cancer cell motility, invasion, and metastasis. However, the mechanism by which ACTN4 drives these malignant phenotypes remains unclear. Here, we show that ACTN4, but not ACTN1, induces the formation of immature focal adhesions in DLD-1 cells, leading to the rapid turnover of focal adhesions. Interestingly, zyxin (ZYX) assembly to focal adhesions was markedly decreased in ACTN4-expressing DLD-1 cells, while the recruitment of paxillin (PAX) occurred normally. On the other hand, in ACTN1-expressing DLD-1 cells, PAX and ZYX were normally recruited to focal adhesions, suggesting that ACTN4 specifically impairs focal adhesion maturation by inhibiting the recruitment of ZYX to focal complexes. Using purified recombinant proteins, we found that ZYX binding to ACTN4 was defective under conditions where ZYX binding to ACTN1 was observed. Furthermore, Matrigel invasion of SW480 cells that express high endogenous levels of ACTN4 protein was inhibited by ectopic expression of ACTN1. Altogether, our results suggest that ZYX defective binding to ACTN4, which occupies focal adhesions instead of ACTN1, induces the formation of immature focal adhesions, resulting in the enhancement of cell motility and invasion.


Subject(s)
Actinin/metabolism , Focal Adhesions/metabolism , Actinin/antagonists & inhibitors , Actinin/genetics , Caco-2 Cells , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , HT29 Cells , Humans , Microscopy, Fluorescence , Neoplasm Invasiveness , Paxillin/metabolism , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Zyxin/chemistry , Zyxin/genetics , Zyxin/metabolism
5.
PLoS One ; 10(4): e0124274, 2015.
Article in English | MEDLINE | ID: mdl-25902045

ABSTRACT

Voltage-dependent L-type calcium channels that permit cellular calcium influx are essential in calcium-mediated modulation of cellular signaling. Although the regulation of voltage-dependent L-type calcium channels is linked to many factors including cAMP-dependent protein kinase A (PKA) activity and actin cytoskeleton, little is known about the detailed mechanisms underlying the regulation in osteoblasts. Our present study investigated the modulation of L-type calcium channel activities through the effects of forskolin on actin reorganization and on its functional interaction with actin binding protein actinin 4. The results showed that forskolin did not significantly affect the trafficking of pore forming α1c subunit and its interaction with actin binding protein actinin 4, whereas it significantly increased the expression of ß3 subunit and its interaction with actinin 4 in osteoblast cells as assessed by co-immunoprecipitation, pull-down assay, and immunostaining. Further mapping showed that the ABD and EF domains of actinin 4 were interaction sites. This interaction is independent of PKA phosphorylation. Knockdown of actinin 4 significantly decreased the activities of L-type calcium channels. Our study revealed a new aspect of the mechanisms by which the forskolin activation of adenylyl cyclase - cAMP cascade regulates the L-type calcium channel in osteoblast cells, besides the PKA mediated phosphorylation of the channel subunits. These data provide insight into the important role of interconnection among adenylyl cyclase, cAMP, PKA, the actin cytoskeleton, and the channel proteins in the regulation of voltage-dependent L-type calcium channels in osteoblast cells.


Subject(s)
Actinin/genetics , Calcium Channels, L-Type/genetics , Calcium/metabolism , Colforsin/pharmacology , Osteoblasts/drug effects , Protein Subunits/genetics , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actinin/antagonists & inhibitors , Actinin/metabolism , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Animals , Calcium Channels, L-Type/metabolism , Calcium Signaling , Cell Line, Tumor , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation , Membrane Potentials/drug effects , Membrane Potentials/physiology , Osteoblasts/cytology , Osteoblasts/metabolism , Patch-Clamp Techniques , Phosphorylation/drug effects , Protein Binding , Protein Subunits/agonists , Protein Subunits/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats
6.
J Biol Chem ; 290(1): 338-49, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25411248

ABSTRACT

Glomerular podocytes are highly specialized terminally differentiated cells that act as a filtration barrier in the kidney. Mutations in the actin-binding protein, α-actinin 4 (ACTN4), are linked to focal segmental glomerulosclerosis (FSGS), a chronic kidney disease characterized by proteinuria. Aberrant activation of NF-κB pathway in podocytes is implicated in glomerular diseases including proteinuria. We demonstrate here that stable knockdown of ACTN4 in podocytes significantly reduces TNFα-mediated induction of NF-κB target genes, including IL-1ß and NPHS1, and activation of an NF-κB-driven reporter without interfering with p65 nuclear translocation. Overexpression of ACTN4 and an actin binding-defective variant increases the reporter activity. In contrast, an FSGS-linked ACTN4 mutant, K255E, which has increased actin binding activity and is predominantly cytoplasmic, fails to potentiate NF-κB activity. Mechanistically, IκBα blocks the association of ACTN4 and p65 in the cytosol. In response to TNFα, both NF-κB subunits p65 and p50 translocate to the nucleus, where they bind and recruit ACTN4 to their targeted promoters, IL-1ß and IL-8. Taken together, our data identify ACTN4 as a novel coactivator for NF-κB transcription factors in podocytes. Importantly, this nuclear function of ACTN4 is independent of its actin binding activity in the cytoplasm.


Subject(s)
Actinin/genetics , NF-kappa B/genetics , Podocytes/metabolism , Transcription, Genetic , Actinin/antagonists & inhibitors , Actinin/metabolism , Actins/genetics , Actins/metabolism , Animals , Cell Line, Transformed , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Podocytes/cytology , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
7.
Cell Mol Life Sci ; 71(22): 4457-70, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24788042

ABSTRACT

Heparanase activity plays a decisive role in cell dissemination associated with cancer metastasis. Cellular uptake of heparanase is considered a pre-requisite for the delivery of latent 65-kDa heparanase to lysosomes and its subsequent proteolytic processing and activation into 8- and 50-kDa protein subunits by cathepsin L. Heparan sulfate proteoglycans, and particularly syndecan, are instrumental for heparanase uptake and activation, through a process that has been shown to occur independent of rafts. Nevertheless, the molecular mechanism underlying syndecan-mediated internalization outside of rafts is unclear. Here, we examined the role of syndecan-1 cytoplasmic domain in heparanase processing, utilizing deletion constructs lacking the entire cytoplasmic domain (Delta), the conserved (C1 or C2), or variable (V) regions. Heparanase processing was markedly increased following syndecan-1 over-expression; in contrast, heparanase was retained at the cell membrane and its processing was impaired in cells over-expressing syndecan-1 deleted for the entire cytoplasmic tail. We have next revealed that conserved domain 2 (C2) and variable (V) regions of syndecan-1 cytoplasmic tail mediate heparanase processing. Furthermore, we found that syntenin, known to interact with syndecan C2 domain, and α actinin are essential for heparanase processing.


Subject(s)
Actinin/metabolism , Glucuronidase/metabolism , Syndecan-1/metabolism , Syntenins/metabolism , Actinin/antagonists & inhibitors , Actinin/genetics , Animals , Cell Line, Tumor , Cell Membrane/metabolism , HEK293 Cells , Humans , Mice , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering/metabolism , Syndecan-1/chemistry , Syndecan-1/genetics , Syntenins/antagonists & inhibitors , Syntenins/genetics
8.
Oncol Rep ; 30(3): 1073-80, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23817592

ABSTRACT

In the present study, the association between clinicopathological parameters and α-actinin-4 (ACTN4) expression in bladder cancer specimens was evaluated, and the functional role of ACTN4 in bladder cancer cells was investigated. Immunohistochemistry using anti-ACTN4 antibody was performed in bladder cancer specimens (53 superficial and 42 muscle-invasive cases) from 95 patients who underwent radical cystectomy (n=46) or transurethral resection (TUR) only (n=49). We divided the levels of ACTN4 expression into 2 groups (low or high) by comparing the staining intensity in each specimen with that of the vascular endothelial cells in the same specimen, and we evaluated the correlations between these levels and pathological parameters, recurrence and prognosis. We also investigated the effects of ACTN4 suppression by siRNA on the invasive ability and proliferation of T24 and KU19-19 cells. High ACTN4 expression was significantly associated with higher tumor grade and higher pT stage. In patients with superficial bladder cancer treated only by TUR, the rate of intravesical recurrence did not differ significantly between patients with high ACTN4 expression and patients with low ACTN4 expression. In patients who had muscle­invasive tumors and underwent radical cystectomy, high ACTN4 expression was associated with neither recurrence nor poor prognosis. Nonetheless, high ACTN4 expression was shown by a large percentage (81%) of patients with muscle-invasive bladder cancer and by a small percentage (17%) of patients with superficial bladder cancer. Furthermore, the leading edges of the invasive bladder cancer showed increased ACTN4 expression. ACTN4 suppression significantly reduced the number of invading bladder cancer cells but unexpectedly increased the proliferation of bladder cancer cells. ACTN4 suppression increased the phosphorylation of ERKs but not AKT or STAT3, suggesting that the increased proliferation due to ACTN4 suppression was mediated in part by the ERK pathway. ACTN4 expression may suppress the proliferation of bladder cancer cells and may produce conditions which facilitate cancer cell invasion.


Subject(s)
Actinin/metabolism , Cell Movement , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Actinin/antagonists & inhibitors , Actinin/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Apoptosis , Blotting, Western , Carcinoma in Situ/metabolism , Carcinoma in Situ/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Adhesion , Cell Proliferation , Collagen/metabolism , Drug Combinations , Female , Humans , Immunoenzyme Techniques , Laminin/metabolism , Male , Middle Aged , Muscle Neoplasms/metabolism , Muscle Neoplasms/pathology , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , Proteoglycans/metabolism , RNA, Small Interfering/genetics , Tumor Cells, Cultured
9.
Oncogene ; 32(4): 462-70, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-22370640

ABSTRACT

Murine double minute (MDM2) binding protein (MTBP) has been implicated in cancer progression. Here, we demonstrate one mechanism by which MTBP inhibits cancer metastasis. Overexpression of MTBP in human osteosarcoma cell lines lacking wild-type p53 did not alter primary tumor growth in mice, but significantly inhibited metastases. MTBP downregulation increased the migratory potential of MDM2(-/-)p53(-/-) mouse embryonic fibroblasts, suggesting that MTBP inhibited cell migration independently of the Mdm2-p53 pathway. Co-immunoprecipitation and mass spectrometric analysis identified alpha-actinin-4 (ACTN4) as an MTBP-interacting protein. Endogenous MTBP interacted with and partially colocalized with ACTN4. MTBP overexpression inhibited cell migration and filopodia formation mediated by ACTN4. Increased cell migration by MTBP downregulation was inhibited by concomitant downregulation of ACTN4. MTBP also inhibited ACTN4-mediated F-actin bundling. We furthermore demonstrated that nuclear localization of MTBP was dispensable for inhibiting ACTN4-mediated cell migration and filopodia formation. Thus, MTBP suppresses cell migration, at least partially, by inhibiting ACTN4 function. Our study not only provides a mechanism of metastasis suppression by MTBP, but also suggests MTBP as a potential biomarker for cancer progression.


Subject(s)
Actinin/antagonists & inhibitors , Actinin/metabolism , Carrier Proteins/metabolism , Cell Movement/physiology , Pseudopodia/metabolism , Actinin/genetics , Actins/genetics , Actins/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Carrier Proteins/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Disease Progression , Down-Regulation , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Pseudopodia/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
10.
FASEB J ; 27(2): 546-56, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23085994

ABSTRACT

During wound repair, epidermal cells at the edge of an injury establish front-rear polarity through orchestrated changes in their cytoskeleton and adhesion structures. The polarity and directed migration of such cells is determined by the assembly, extension, and stabilization of a lamellipodium. Actinin-4 associates with lamellipodia and has been implicated in regulating lamellipodial structure, function and assembly. To study the functions of actinin-4 in human keratinocytes, we used shRNA to generate knockdown cells and compared their motility behavior and matrix adhesion assembly to scrambled shRNA treated control keratinocytes. Actinin-4 knockdown keratinocytes lack polarity, assemble multiple lamellipodia with a 2× increased area over controls, display reduced activity of the actin remodeling protein cofilin, and fail to migrate in a directional manner. This motility defect is rescued by plating knockdown cells on preformed laminin-332 matrix. In actinin-4-knockdown keratinocytes, focal contact area is increased by 25%, and hemidesmosome proteins are mislocalized. Specifically, α6ß4 integrin localizes to large lamellipodial extensions, displays reduced dynamics, and fails to recruit its bullous pemphigoid antigen binding partners. Together, our data indicate a role for actinin-4 in regulating the steering mechanism of keratinocytes via profound effects on their matrix adhesion sites.


Subject(s)
Actinin/physiology , Keratinocytes/physiology , Pseudopodia/physiology , Actin Depolymerizing Factors/physiology , Actinin/antagonists & inhibitors , Actinin/genetics , Cell Movement/physiology , Cells, Cultured , Focal Adhesions/physiology , Gene Knockdown Techniques , Hemidesmosomes/physiology , Humans , Integrin alpha6beta4/genetics , Integrin alpha6beta4/physiology , RNA, Small Interfering/genetics
11.
Cancer Metastasis Rev ; 31(3-4): 633-40, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22684342

ABSTRACT

MDM2 binding protein (MTBP) is a protein that interacts with oncoprotein murine double minute (MDM2), a major inhibitor of the tumor suppressor p53. Overexpression of MTBP leads to p53-independent cell proliferation arrest, which is in turn blocked by simultaneous overexpression of MDM2. Importantly, reduced expression of MTBP in mice increases tumor metastasis and enhances migratory potential of mouse embryonic fibroblasts regardless of the presence of p53. Clinically, loss of MTBP expression in head and neck squamous cell carcinoma is associated with reduced patient survival, and is shown to serve as an independent prognostic factor when p53 is mutated in tumors. These results indicate the involvement of MTBP in suppressing tumor progression. Our recent findings demonstrate that overexpression of MTBP in human osteosarcoma cells lacking wild-type p53 inhibits metastasis, but not primary tumor growth, when cells are transplanted in femurs of immunocompromised mice. These data indicate that MTBP functions as a metastasis suppressor independent of p53 status. Furthermore, overexpression of MTBP suppresses cell migration and filopodia formation, in part, by inhibiting function of an actin crosslinking protein α-actinin-4. Thus, increasing evidence indicates the significance of MTBP in tumor progression. We summarize published results related to MTBP function and discuss caveats and future directions in this review article.


Subject(s)
Carrier Proteins/physiology , Neoplasm Metastasis/prevention & control , Tumor Suppressor Proteins/physiology , Actinin/antagonists & inhibitors , Actinin/physiology , Animals , Carrier Proteins/chemistry , Cell Movement , Humans , Mice , Tumor Suppressor Protein p53/physiology
12.
Cell Cycle ; 11(10): 1929-37, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22544326

ABSTRACT

The actin cross-linking protein, α-actinin, plays a crucial role in mediating furrow ingression during cytokinesis. However, the mechanism by which its dynamics are regulated during this process is poorly understood. Here we have investigated the role of calcium sensitivity of α-actinin in the regulation of its dynamics by generating a functional calcium-insensitive mutant (EFM). GFP-tagged EFM (EFM-GFP) localized to the equatorial regions during cell division. However, the maximal equatorial accumulation of EFM-GFP was significantly smaller in comparison to α-actinin-GFP when it was expressed in normal cells and cells depleted of endogenous α-actinin. No apparent defects in cytokinesis were observed in these cells. However, F-actin levels at the equator were significantly reduced in cells expressing EFM-GFP as compared with α-actinin-GFP at furrow initiation but were recovered during furrow ingression. These results suggest that calcium sensitivity of α-actinin is required for its equatorial accumulation that is crucial for the initial equatorial actin assembly but is dispensable for cytokinesis. Equatorial RhoA localization was not affected by EFM-GFP overexpression, suggesting that equatorial actin assembly is predominantly driven by the RhoA-dependent mechanism. Our observations shed new light on the role and regulation of the accumulation of pre-existing actin filaments in equatorial actin assembly during cytokinesis.


Subject(s)
Actinin/metabolism , Calcium/metabolism , Cytokinesis/physiology , Actinin/antagonists & inhibitors , Actinin/genetics , Actins/metabolism , Amino Acid Sequence , Animals , Cell Line , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mutation , RNA Interference , RNA, Small Interfering , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
13.
Oncol Rep ; 25(3): 887-93, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21234524

ABSTRACT

Invasion of normal brain tissue by brain tumor cells is a major contributing factor to the recurrence and resistance of clinically diagnosed glioblastomas to therapy (surgery, chemotherapy, radiation). Here, we have assessed the efficacy of the microtubule inhibiting agent epothilone B on glioblastoma cell motility, a prerequisite cellular program of invasive glioblastomas. Using cell migration assays and immunofluorescence techniques we demonstrated that epothilone B abrogated glioblastoma cell motility as a consequence of α-actinin 4 redistristrubiton and the breakdown of cellular structures (leading edge, stress fibers) it is associated with during cell migration. Evaluation of the microtubule actin cross linking factor in glioblastoma cells also revealed epothilone B invoked changes in this cytoskeleton cross linking protein, resembling α-actinin 4 changes in response to epothilone B. We have demonstrated in this study that epothilone B antagonizes glioblastoma cell motility due to the disruption of cytoskeleton binding proteins that aide in preserving the structural organization of the cytoskeleton filamentous network. Furthermore, we provide preclincial evidence that epothilone B effects on glioblastomas are not limited to the impairment of dividing tumors cells but that it also targets migratory and invasive glioblastoma cells, suggesting that this agent has potential clinical benefit due to its ability to target divergent cellular programs in the glioblastoma tumor mass.


Subject(s)
Actinin/antagonists & inhibitors , Brain Neoplasms/pathology , Cell Movement/drug effects , Epothilones/pharmacology , Glioma/pathology , Tubulin Modulators/pharmacology , Actinin/metabolism , Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Down-Regulation , Drug Evaluation, Preclinical , Glioma/metabolism , Humans , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/metabolism , Microtubules/drug effects , Microtubules/metabolism , Protein Multimerization/drug effects , Protein Transport/drug effects , Tumor Cells, Cultured
14.
J Mol Biol ; 403(4): 516-28, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20850453

ABSTRACT

The actin binding protein α-actinin is a major component of focal adhesions found in vertebrate cells and of focal-adhesion-like structures found in the body wall muscle of the nematode Caenorhabditis elegans. To study its in vivo function in this genetic model system, we isolated a strain carrying a deletion of the single C. elegans α-actinin gene. We assessed the cytological organization of other C. elegans focal adhesion proteins and the ultrastructure of the mutant. The mutant does not have normal dense bodies, as observed by electron microscopy; however, these dense-body-like structures still contain the focal adhesion proteins integrin, talin, and vinculin, as observed by immunofluorescence microscopy. Actin is found in normal-appearing I-bands, but with abnormal accumulations near muscle cell membranes. Although swimming in water appeared grossly normal, use of automated methods for tracking the locomotion of individual worms revealed a defect in bending. We propose that the reduced motility of α-actinin null is due to abnormal dense bodies that are less able to transmit the forces generated by actin/myosin interactions.


Subject(s)
Actinin/physiology , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Focal Adhesions/physiology , Actinin/antagonists & inhibitors , Actinin/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Focal Adhesions/genetics , Gene Deletion , Genes, Helminth , Locomotion/physiology , Microscopy, Fluorescence , Muscles/physiology , Mutation , Phenotype , RNA Interference
15.
Bioorg Med Chem ; 17(3): 977-80, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-18329275

ABSTRACT

A small molecule inhibitor of alpha4 integrin-dependent cell migration was identified through a cell-based screen of small molecule libraries. Biochemical and cellular experiments suggest that this molecule functions by interacting with gamma-parvin. This molecule should serve as a useful tool to study alpha4 integrin signaling and may lead to new therapeutics for the treatment of autoimmune diseases.


Subject(s)
Aniline Compounds/pharmacology , Cell Movement/drug effects , Integrin alpha4/metabolism , Tubercidin/analogs & derivatives , Actinin/antagonists & inhibitors , Actinin/metabolism , Adenosine Kinase/antagonists & inhibitors , Adenosine Kinase/metabolism , Aniline Compounds/chemical synthesis , Aniline Compounds/chemistry , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Humans , Integrin alpha4/drug effects , Jurkat Cells , RNA Interference , Signal Transduction , Small Molecule Libraries , Tubercidin/chemical synthesis , Tubercidin/chemistry , Tubercidin/pharmacology
16.
Clin Cancer Res ; 14(17): 5348-56, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18765526

ABSTRACT

PURPOSE: An invasive growth pattern is one of the hallmarks of pancreatic ductal carcinoma. Actinin-4 is an actin-binding protein associated with enhanced cell motility, invasive growth, and lymph node metastasis. Actinin-4 might play an important role in the development and progression of pancreatic cancer. EXPERIMENTAL DESIGN: The expression of actinin-4 was examined immunohistochemically in 173 cases of invasive pancreatic ductal carcinoma. The copy number of the actinin-4 (ACTN4) gene was calculated by fluorescence in situ hybridization. The expression of actinin-4 was stably knocked down by short hairpin RNA, and tumorigenicity was evaluated by orthotopic implantation into mice with severe combined immunodeficiency. RESULTS: The expression level of actinin-4 was increased in 109 (63.0%) of 173 cases of pancreatic cancer. Kaplan-Meier survival curves revealed that patients with increased expression of actinin-4 had a significantly poorer outcome (P=0.00001, log-rank test). Multivariate analysis by the Cox proportional hazard model showed that high expression of actinin-4 was the most significant independent negative predictor of survival (hazard ratio, 2.33; P=0.000009). Amplification (defined as more than four copies per interphase nucleus) of the ACTN4 gene was detected in 11 (37.9%) of 29 cases showing increased expression of actinin-4. Knockdown of actinin-4 expression inhibited the destructive growth of cancer cells in the pancreatic parenchyma. CONCLUSION: Recurrent amplification of chromosome 19q13.1-2 has been reported in pancreatic cancer, but the exact target gene has not been identified. Actinin-4 contributes to the invasive growth of pancreatic ductal carcinoma, and ACTN4 is one of the candidate oncogenes in this chromosome locus.


Subject(s)
Actinin/genetics , Carcinoma, Pancreatic Ductal/genetics , Gene Amplification , Pancreatic Neoplasms/genetics , Actinin/antagonists & inhibitors , Aged , Animals , Cell Line, Tumor , Female , Gene Expression , Humans , Male , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation , Transfection
17.
Cell Mol Life Sci ; 65(9): 1446-57, 2008 May.
Article in English | MEDLINE | ID: mdl-18392556

ABSTRACT

Physical forces can activate colon cancer cell adhesion, critical for metastasis. Paxillin is phosphorylated by FAK and required for pressure-stimulated adhesion. However, whether paxillin acts as an inert scaffolding protein or whether paxillin phosphorylation is required is unknown. Transfection with paxillin point-phosphorylation mutants demonstrated that phosphorylation at tyrosines 31 and 118 together is necessary for pressure-stimulated adhesion. We further evaluated potential paxillin partners. Reducing the adaptor protein Crk or the focal adhesion protein p130Cas blocked pressure-stimulated adhesion. Furthermore, Crk and p130Cas both displayed increased co-immunoprecipitation with paxillin in response to increased pressure, except in cells transfected with a Y31Y118 paxillin mutant. Inhibiting the small GTPase Rac1 also abolished pressure-stimulated adhesion, and reducing paxillin by siRNA blocked Rac1 phosphorylation by pressure. Thus, paxillin phosphorylation at tyrosines 31 and 118 together is necessary for pressure-induced adhesion. Paxillin, Crk and Cas form a trimeric complex that activates Rac1 and mediates this effect.


Subject(s)
Colonic Neoplasms/metabolism , Crk-Associated Substrate Protein/metabolism , Paxillin/metabolism , Proto-Oncogene Proteins c-crk/metabolism , rac1 GTP-Binding Protein/metabolism , Actinin/antagonists & inhibitors , Cell Adhesion , Cell Line, Tumor , Colonic Neoplasms/pathology , Crk-Associated Substrate Protein/antagonists & inhibitors , Humans , Neoplasm Metastasis , Paxillin/antagonists & inhibitors , Paxillin/chemistry , Phosphorylation , Pressure , Proto-Oncogene Proteins c-crk/antagonists & inhibitors , RNA Interference , Tumor Cells, Cultured , Tyrosine/metabolism
18.
Am J Physiol Cell Physiol ; 291(5): C909-21, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16807302

ABSTRACT

Maintenance of bone structural integrity depends in part on the rate of apoptosis of bone-forming osteoblasts. Because substrate adhesion is an important regulator of apoptosis, we have investigated the role of focal adhesions in regulating bone cell apoptosis. To test this, we expressed a truncated form of alpha-actinin (ROD-GFP) that competitively displaces endogenous alpha-actinin from focal adhesions, thus disrupting focal adhesions. Immunofluorescence and morphometric analysis of vinculin and tyrosine phosphorylation revealed that ROD-GFP expression dramatically disrupted focal adhesion organization and reduced tyrosine phosphorylation at focal adhesions. In addition, Bcl-2 protein levels were reduced in ROD-GFP-expressing cells, but caspase 3 cleavage, poly(ADP-ribose) polymerase cleavage, histone H2A.X phosphorylation, and cytotoxicity were not increased due to ROD-GFP expression alone. Increases in both ERK and Akt phosphorylation were also observed in ROD-GFP-expressing cells, although inhibition of either ERK or Akt individually or together failed to induce apoptosis. However, we did find that ROD-GFP expression sensitized, whereas alpha-actinin-GFP expression protected, cells from TNF-alpha-induced apoptosis. Further investigation revealed that activation of TNF-alpha-induced survival signals, specifically Akt phosphorylation and NF-kappaB activation, was inhibited in ROD-GFP-expressing cells. The reduced expression of antiapoptotic Bcl-2 and inhibited survival signaling rendered ROD-GFP-expressing cells more susceptible to TNF-alpha-induced apoptosis. Thus we conclude that alpha-actinin plays a role in regulating cell survival through stabilization of focal adhesions and regulation of TNF-alpha-induced survival signaling.


Subject(s)
Actinin/metabolism , Apoptosis/physiology , Focal Adhesions/metabolism , Integrins/metabolism , Mutant Proteins/metabolism , Osteoblasts/cytology , Actinin/antagonists & inhibitors , Actins/metabolism , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesions/drug effects , Humans , Integrins/antagonists & inhibitors , Mice , NF-kappa B/metabolism , Osteoblasts/drug effects , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/metabolism , Stress Fibers/drug effects , Stress Fibers/metabolism , Tumor Necrosis Factor-alpha/pharmacology
19.
J Biol Chem ; 278(26): 24039-45, 2003 Jun 27.
Article in English | MEDLINE | ID: mdl-12716899

ABSTRACT

alpha-Actinin is an abundant actin-bundling and adhesion protein that directly links actin filaments to integrin receptors. Previously, in platelet-derived growth factor-treated fibroblasts, we demonstrated that phosphoinositides bind to alpha-actinin, regulating its localization (Greenwood, J. A., Theibert, A. B., Prestwich, G. D., and Murphy-Ullrich, J. E. (2000) J. Cell Biol. 150, 627- 642). In this study, phosphoinositide binding and regulation of alpha-actinin function is further characterized. Phosphoinositide binding specificity, determined using a protein-lipid overlay procedure, suggests that alpha-actinin interacts with phosphates on the 4th and 5th position of the inositol head group. Binding assays and mutational analyses demonstrate that phosphoinositides bind to the calponin homology domain 2 of alpha-actinin. Phosphoinositide binding inhibited the bundling activity of alpha-actinin by blocking the interaction of the actin-binding domain with actin filaments. Consistent with these results, excessive bundling of actin filaments was observed in fibroblasts expressing an alpha-actinin mutant with decreased phosphoinositide affinity. We conclude that the interaction of alpha-actinin with phosphoinositides regulates actin stress fibers in the cell by controlling the extent to which microfilaments are bundled.


Subject(s)
Actinin/antagonists & inhibitors , Actinin/physiology , Phosphatidylinositols/pharmacology , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Binding Sites , Chickens , Dose-Response Relationship, Drug , Fibroblasts/chemistry , Mutation , Protein Binding , Rats , Stress Fibers/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...