1
|
Gholam Azad M, Hussaini M, Russell TM, Richardson V, Kaya B, Dharmasivam M, Richardson DR. Multi-modal mechanisms of the metastasis suppressor, NDRG1: Inhibition of WNT/β-catenin signaling by stabilization of protein kinase Cα. J Biol Chem 2024; 300:107417. [PMID: 38815861 PMCID: PMC11261793 DOI: 10.1016/j.jbc.2024.107417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 06/01/2024] Open
Abstract
The metastasis suppressor, N-myc downstream regulated gene-1 (NDRG1), inhibits pro-oncogenic signaling in pancreatic cancer (PC). This investigation dissected a novel mechanism induced by NDRG1 on WNT/β-catenin signaling in multiple PC cell types. NDRG1 overexpression decreased β-catenin and downregulated glycogen synthase kinase-3β (GSK-3β) protein levels and its activation. However, β-catenin phosphorylation at Ser33, Ser37, and Thr41 are classically induced by GSK-3β was significantly increased after NDRG1 overexpression, suggesting a GSK-3β-independent mechanism. Intriguingly, NDRG1 overexpression upregulated protein kinase Cα (PKCα), with PKCα silencing preventing β-catenin phosphorylation at Ser33, Ser37, and Thr41, and decreasing β-catenin expression. Further, NDRG1 and PKCα were demonstrated to associate, with PKCα stabilization occurring after NDRG1 overexpression. PKCα half-life increased from 1.5 ± 0.8 h (3) in control cells to 11.0 ± 2.5 h (3) after NDRG1 overexpression. Thus, NDRG1 overexpression leads to the association of NDRG1 with PKCα and PKCα stabilization, resulting in β-catenin phosphorylation at Ser33, Ser37, and Thr41. The association between PKCα, NDRG1, and β-catenin was identified, with the formation of a potential metabolon that promotes the latter β-catenin phosphorylation. This anti-oncogenic activity of NDRG1 was multi-modal, with the above mechanism accompanied by the downregulation of the nucleo-cytoplasmic shuttling protein, p21-activated kinase 4 (PAK4), which is involved in β-catenin nuclear translocation, inhibition of AKT phosphorylation (Ser473), and decreased β-catenin phosphorylation at Ser552 that suppresses its transcriptional activity. These mechanisms of NDRG1 activity are important to dissect to understand the marked anti-cancer efficacy of NDRG1-inducing thiosemicarbazones that upregulate PKCα and inhibit WNT signaling.
Collapse
Affiliation(s)
- Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Mohammed Hussaini
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Tiffany M Russell
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Vera Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
2
|
Garg A, Bandyopadhyay S. Role of an interdependent Wnt, GSK3-β/β-catenin and HB-EGF/EGFR mechanism in arsenic-induced hippocampal neurotoxicity in adult mice. CHEMOSPHERE 2024; 352:141375. [PMID: 38325618 DOI: 10.1016/j.chemosphere.2024.141375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/17/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
We previously reported the neurotoxic effects of arsenic in the hippocampus. Here, we explored the involvement of Wnt pathway, which contributes to neuronal functions. Administering environmentally relevant arsenic concentrations to postnatal day-60 (PND60) mice demonstrated a dose-dependent increase in hippocampal Wnt3a and its components, Frizzled, phospho-LRP6, Dishevelled and Axin1 at PND90 and PND120. However, p-GSK3-β(Ser9) and β-catenin levels although elevated at PND90, decreased at PND120. Additionally, treatment with Wnt-inhibitor, rDkk1, reduced p-GSK3-β(Ser9) and β-catenin at PND90, but failed to affect their levels at PND120, indicating a time-dependent link with Wnt. To explore other underlying factors, we assessed epidermal growth factor receptor (EGFR) pathway, which interacts with GSK3-β and appears relevant to neuronal functions. We primarily found that arsenic reduced hippocampal phosphorylated-EGFR and its ligand, Heparin-binding EGF-like growth factor (HB-EGF), at both PND90 and PND120. Moreover, treatment with HB-EGF rescued p-GSK3-β(Ser9) and β-catenin levels at PND120, suggesting their HB-EGF/EGFR-dependent regulation at this time point. Additionally, rDkk1, LiCl (GSK3-β-activity inhibitor), or β-catenin protein treatments induced a time-dependent recovery in HB-EGF, indicating potential inter-dependent mechanism between hippocampal Wnt/β-catenin and HB-EGF/EGFR following arsenic exposure. Fluorescence immunolabeling then validated these findings in hippocampal neurons. Further exploration of hippocampal neuronal survival and apoptosis demonstrated that treatment with rDkk1, LiCl, β-catenin and HB-EGF improved Nissl staining and NeuN levels, and reduced cleaved-caspase-3 levels in arsenic-treated mice. Supportively, we detected improved Y-Maze and Passive Avoidance performances for learning-memory functions in these mice. Overall, our study provides novel insights into Wnt/β-catenin and HB-EGF/EGFR pathway interaction in arsenic-induced hippocampal neurotoxicity.
Collapse
Affiliation(s)
- Asmita Garg
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sanghamitra Bandyopadhyay
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
3
|
Young IC, Brabletz T, Lindley LE, Abreu M, Nagathihalli N, Zaika A, Briegel KJ. Multi-cancer analysis reveals universal association of oncogenic LBH expression with DNA hypomethylation and WNT-Integrin signaling pathways. Cancer Gene Ther 2023; 30:1234-1248. [PMID: 37268816 PMCID: PMC10501907 DOI: 10.1038/s41417-023-00633-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 05/08/2023] [Accepted: 05/19/2023] [Indexed: 06/04/2023]
Abstract
Limb-Bud and Heart (LBH) is a developmental transcription co-factor deregulated in cancer, with reported oncogenic and tumor suppressive effects. However, LBH expression in most cancer types remains unknown, impeding understanding of its mechanistic function Here, we performed systematic bioinformatic and TMA analysis for LBH in >20 different cancer types. LBH was overexpressed in most cancers compared to normal tissues (>1.5-fold; p < 0.05), including colon-rectal, pancreatic, esophageal, liver, stomach, bladder, kidney, prostate, testicular, brain, head & neck cancers, and sarcoma, correlating with poor prognosis. The cancer types showing LBH downregulation were lung, melanoma, ovarian, cervical, and uterine cancer, while both LBH over- and under-expression were observed in hematopoietic malignancies. In cancers with LBH overexpression, the LBH locus was frequently hypomethylated, identifying DNA hypomethylation as a potential mechanism for LBH dysregulation. Pathway analysis identified a universal, prognostically significant correlation between LBH overexpression and the WNT-Integrin signaling pathways. Validation of the clinical association of LBH with WNT activation in gastrointestinal cancer cell lines, and in colorectal patient samples by IHC uncovered that LBH is specifically expressed in tumor cells with nuclear beta-catenin at the invasive front. Collectively, these data reveal a high degree of LBH dysregulation in cancer and establish LBH as pan-cancer biomarker for detecting WNT hyperactivation in clinical specimens.
Collapse
Affiliation(s)
- In-Chi Young
- Department of Surgery, Division of Surgical Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Linsey E Lindley
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
- Graduate Program in Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Maria Abreu
- Department of Medicine, Division of Gastroenterology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nagaraj Nagathihalli
- Department of Surgery, Division of Surgical Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alexander Zaika
- Department of Surgery, Division of Surgical Oncology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Karoline J Briegel
- Department of Surgery, Division of Surgical Oncology, University of Miami Miller School of Medicine, Miami, FL, USA.
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
4
|
Tabassum S, Basu M, Ghosh MK. The DEAD-box RNA helicase DDX5 (p68) and β-catenin: The crucial regulators of FOXM1 gene expression in arbitrating colorectal cancer. BIOCHIMICA ET BIOPHYSICA ACTA (BBA) - GENE REGULATORY MECHANISMS 2023; 1866:194933. [PMID: 36997114 DOI: 10.1016/j.bbagrm.2023.194933] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/23/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023]
Abstract
Forkhead box M1 (FOXM1), a vital member of the Forkhead box family of transcription factors, helps in mediating oncogenesis. However, limited knowledge exists regarding the mechanistic insights into the FOXM1 gene regulation. DDX5 (p68), an archetypal member of the DEAD-box family of RNA helicases, shows multifaceted action in cancer progression by arbitrating RNA metabolism and transcriptionally coactivating transcription factors. Here, we report a novel mechanism of alliance between DDX5 (p68) and the Wnt/β-catenin pathway in regulating FOXM1 gene expression and driving colon carcinogenesis. Initial bioinformatic analyses highlighted elevated expression levels of FOXM1 and DDX5 (p68) in colorectal cancer datasets. Immunohistochemical assays confirmed that FOXM1 showed a positive correlation with DDX5 (p68) and β-catenin in both normal and colon carcinoma patient samples. Overexpression of DDX5 (p68) and β-catenin increased the protein and mRNA expression profiles of FOXM1, and the converse correlation occurred during downregulation. Mechanistically, overexpression and knockdown of DDX5 (p68) and β-catenin elevated and diminished FOXM1 promoter activity respectively. Additionally, Chromatin immunoprecipitation assay demonstrated the occupancy of DDX5 (p68) and β-catenin at the TCF4/LEF binding element (TBE) sites on the FOXM1 promoter. Thiostrepton delineated the effect of FOXM1 inhibition on cell proliferation and migration. Colony formation assay, migration assay, and cell cycle data reveal the importance of the DDX5 (p68)/β-catenin/FOXM1 axis in oncogenesis. Collectively, our study mechanistically highlights the regulation of FOXM1 gene expression by DDX5 (p68) and β-catenin in colorectal cancer.
Collapse
|
5
|
Park SJ, Kim JH, Oh S, Lee DY. Metabolome-Wide Reprogramming Modulated by Wnt/β-Catenin Signaling Pathway. J Microbiol Biotechnol 2023; 33:114-122. [PMID: 36474320 PMCID: PMC9895996 DOI: 10.4014/jmb.2211.11013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022]
Abstract
A family of signal transduction pathways known as wingless type (Wnt) signaling pathways is essential to developmental processes like cell division and proliferation. Mutation in Wnt signaling results in a variety of diseases, including cancers of the breast, colon, and skin, metabolic disease, and neurodegenerative disease; thus, the Wnt signaling pathways have been attractive targets for disease treatment. However, the complicatedness and large involveness of the pathway often hampers pinpointing the specific targets of the metabolic process. In our current study, we investigated the differential metabolic regulation by the overexpression of the Wnt signaling pathway in a timely-resolved manner by applying high-throughput and un-targeted metabolite profiling. We have detected and annotated 321 metabolite peaks from a total of 36 human embryonic kidney (HEK) 293 cells using GC-TOF MS and LC-Orbitrap MS. The un-targeted metabolomic analysis identified the radical reprogramming of a range of central carbon/nitrogen metabolism pathways, including glycolysis, TCA cycle, and glutaminolysis, and fatty acid pathways. The investigation, combined with targeted mRNA profiles, elucidated an explicit understanding of activated fatty acid metabolism (β-oxidation and biosynthesis). The findings proposed detailed mechanistic biochemical dynamics in response to Wnt-driven metabolic changes, which may help design precise therapeutic targets for Wnt-related diseases.
Collapse
Affiliation(s)
- Soo Jin Park
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Joo-Hyun Kim
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea
| | - Sangtaek Oh
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea,Department of Interdisciplinary Program for Bio-Health Convergence, Kookmin University, Seoul 02707, Republic of Korea,Corresponding authors S. Oh Phone: +82-2-910-5732 E-mail:
| | - Do Yup Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea,Interdisciplinary Program in Agricultural Genomics, Seoul National University, Seoul 08826, Republic of Korea,Center for Food and Bioconvergence, Research Institute for Agricultural and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea,D.Y. Lee Phone: + 82-2-880-5644 E-mail:
| |
Collapse
|
6
|
Condello S, Prasad M, Atwani R, Matei D. Tissue transglutaminase activates integrin-linked kinase and β-catenin in ovarian cancer. J Biol Chem 2022; 298:102242. [PMID: 35810788 PMCID: PMC9358478 DOI: 10.1016/j.jbc.2022.102242] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 10/26/2022] Open
Abstract
Ovarian cancer (OC) is the most lethal gynecological cancer. OC cells have high proliferative capacity, are invasive, resist apoptosis, and tumors often display rearrangement of extracellular matrix (ECM) components, contributing to accelerated tumor progression. The multifunctional protein tissue transglutaminase (TG2) is known to be secreted in the tumor microenvironment (TME), where it interacts with fibronectin (FN) and the cell surface receptor β1 integrin. However, the mechanistic role of TG2 in cancer cell proliferation is unknown. Here, we demonstrate TG2 directly interacts with and facilitates the phosphorylation and activation of the integrin effector protein integrin-linked kinase (ILK) at Ser246. We show TG2 and p-Ser246-ILK form a complex that is detectable in patient-derived OC primary cells grown on FN-coated slides. In addition, we show co-expression of TGM2 and ILK correlates with poor clinical outcome. Mechanistically, we demonstrate TG2-mediated ILK activation causes phosphorylation of glycogen synthase kinase-3α/β (GSK-3α/β), allowing β-catenin nuclear translocation and transcriptional activity. Furthermore, inhibition of TG2 and ILK using small molecules, neutralizing antibodies, or shRNA-mediated knockdown block cell adhesion to the FN matrix, as well as the Wnt receptor response to the Wnt-3A ligand, and ultimately, cell adhesion, growth, and migration. In conclusion, we demonstrate TG2 directly interacts with and activates ILK in OC cells and tumors, and define a new mechanism which links ECM cues with β-catenin signaling in OC. These results suggest a central role of TG2/FN/integrin clusters in ECM rearrangement and indicate downstream effector ILK may represent a potential new therapeutic target in OC.
Collapse
Affiliation(s)
- Salvatore Condello
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202; Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202.
| | - Mayuri Prasad
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202; Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Rula Atwani
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202; Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Daniela Matei
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611; Robert H Lurie Comprehensive Cancer Center, Chicago, IL, USA; Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
7
|
Kal S, Chakraborty S, Karmakar S, Ghosh MK. Wnt/β-catenin signaling and p68 conjointly regulate CHIP in colorectal carcinoma. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119185. [PMID: 34890713 DOI: 10.1016/j.bbamcr.2021.119185] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/23/2021] [Accepted: 11/28/2021] [Indexed: 12/12/2022]
Abstract
Emerging evidences suggest abundant expression of Carboxy terminus of Hsc70 Interacting Protein or CHIP (alias STIP1 Homology and U-box Containing Protein 1 or STUB1) in colorectal carcinoma, but the mechanistic detail of this augmented expression pattern is unclear. The signature driver of canonical Wnt pathway, β-catenin, and its co-activator RNA helicase p68, are also overexpressed in colorectal carcinoma. In this study, we describe a novel mechanism of Wnt/β-catenin and p68 mediated transcriptional activation of CHIP gene leading to enhanced proliferation of colorectal carcinoma cells. Bioinformatic analyses reconfirmed an elevated CHIP expression level in colorectal carcinoma datasets. Wnt3A treatment and pharmacological activation of canonical Wnt signaling pathway resulted in increased nuclear translocation of β-catenin, augmenting CHIP expression. Likewise, immunoblotting and Real time PCR following overexpression and knockdown of β-catenin and p68 demonstrated upregulated and downregulated CHIP expression, respectively, at both mRNA and protein levels. p68 along with β-catenin were found to occupy Transcription Factor 4 (TCF4) binding sites on endogenous CHIP promoter and regulate its transcription. After cloning CHIP promoter, the increased and decreased promoter activities of CHIP induced by overexpression and knockdown of either β-catenin or p68 further confirmed transcriptional regulation of CHIP gene by Wnt/β-catenin signaling cascade. Finally, enhanced cellular propagation and migration of colorectal carcinoma cells induced by 'Wnt/β-catenin-p68-CHIP' axis established the significance of this pathway in oncogenesis. To the best of our knowledge, this is the first report elucidating the mechanistic details of transcriptional regulation of CHIP (STUB1) gene expression.
Collapse
Affiliation(s)
- Satadeepa Kal
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata- 700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Shrabastee Chakraborty
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata- 700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Subhajit Karmakar
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata- 700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Mrinal K Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata- 700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India.
| |
Collapse
|
8
|
Ghazi A, Le Corre D, Pilati C, Taieb J, Aparicio T, Didelot A, Dedhar S, Mulot C, Le Malicot K, Djouadi F, de Reynies A, Launay JM, Laurent-Puig P, Mouillet-Richard S. Prognostic value of the PrP C-ILK-IDO1 axis in the mesenchymal colorectal cancer subtype. Oncoimmunology 2021; 10:1940674. [PMID: 34249475 PMCID: PMC8244775 DOI: 10.1080/2162402x.2021.1940674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
The CMS4 mesenchymal subtype of colorectal cancer (CRC) is associated with poor prognosis and resistance to treatment. The cellular prion protein PrPC is overexpressed in CMS4 tumors and controls the expression of a panel of CMS4-specific genes in CRC cell lines. Here, we sought to investigate PrPC downstream pathways that may underlie its role in CMS4 CRC. By combining gene set enrichment analyses and gain and loss of function approaches in CRC cell lines, we identify the integrin-linked kinase ILK as a proximal effector of PrPC that mediates its control on the CMS4 phenotype. We further leveraged three independent large CRC cohorts to assess correlations in gene expression pattern with patient outcomes and found that ILK is overexpressed in CMS4 mesenchymal tumors and confers a poor prognosis, especially when combined with high expression of the PrPC encoding gene PRNP. Of note, we discovered that the PrPC-ILK signaling axis controls the expression and activity of the tryptophan metabolizing enzyme indoleamine 2,3 dioxygenase IDO1, a key player in immune tolerance. In addition, we monitored alterations in the levels of tryptophan and its metabolites of the kynurenine pathway in the plasma of metastatic CRC patients (n = 325) and we highlight their prognostic value in combination with plasma PrPC levels. Thus, the PrPC-ILK-IDO1 axis plays a key role in the mesenchymal subtype of CRC. PrPC and IDO1-targeted strategies may represent new avenues for patient stratification and treatment in CRC.
Collapse
Affiliation(s)
- Alexandre Ghazi
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Delphine Le Corre
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Camilla Pilati
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Julien Taieb
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France.,Department of Gastroenterology and GI Oncology, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Thomas Aparicio
- Department of Gastroenterology and Digestive Oncology, AP-HP, Hôpital Saint-Louis, Université de Paris, Université Paris Diderot, Paris, France
| | - Audrey Didelot
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Shoukat Dedhar
- Genetics Unit, Integrative Oncology, BC Cancer, Vancouver, Canada
| | - Claire Mulot
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Karine Le Malicot
- Fédération Francophone de Cancérologie Digestive, Epicad Inserm, Université de Bourgogne et and Franche Comté, Dijon, France
| | - Fatima Djouadi
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Aurélien de Reynies
- Programme carte d'identité des tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Jean-Marie Launay
- AP-HP Service de Biochimie, INSERM U942 Lariboisière Hospital, Paris, France.,Pharma Research Department, F. Hoffmann-La-Roche Ltd., Basel, Switzerland
| | - Pierre Laurent-Puig
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France.,Department of Biology, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Sophie Mouillet-Richard
- Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| |
Collapse
|
9
|
Yuzuriha A, Nakamura S, Sugimoto N, Kihara S, Nakagawa M, Yamamoto T, Sekiguchi K, Eto K. Extracellular laminin regulates hematopoietic potential of pluripotent stem cells through integrin β1-ILK-β-catenin-JUN axis. Stem Cell Res 2021; 53:102287. [PMID: 33813173 DOI: 10.1016/j.scr.2021.102287] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 12/21/2022] Open
Abstract
Recombinant matrices have enabled feeder cell-free maintenance cultures of human pluripotent stem cells (hPSCs), with laminin 511-E8 fragment (LM511-E8) being widely used. However, we herein report that hPSCs maintained on LM511-E8 resist differentiating to multipotent hematopoietic progenitor cells (HPCs), unlike hPSCs maintained on LM421-E8 or LM121-E8. The latter two LM-E8s bound weakly to hPSCs compared with LM511-E8 and activated the canonical Wnt/β-catenin signaling pathway. Moreover, the extracellular LM-E8-dependent preferential hematopoiesis was associated with a higher expression of integrin β1 (ITGB1) and downstream integrin-linked protein kinase (ILK), β-catenin and phosphorylated JUN. Accordingly, the lower coating concentration of LM511-E8 or addition of a Wnt/β-catenin signaling activator, CHIR99021, facilitated higher HPC yield. In contrast, the inhibition of ILK, Wnt or JNK by inhibitors or mRNA knockdown suppressed the HPC yield. These findings suggest that extracellular laminin scaffolds modulate the hematopoietic differentiation potential of hPSCs by activating the ITGB1-ILK-β-catenin-JUN axis at the undifferentiated stage. Finally, the combination of low-concentrated LM511-E8 and a revised hPSC-sac method, which adds bFGF, SB431542 and heparin to the conventional method, enabled a higher yield of HPCs and higher rate for definitive hematopoiesis, suggesting a useful protocol for obtaining differentiated hematopoietic cells from hPSCs in general.
Collapse
Affiliation(s)
- Akinori Yuzuriha
- Department of Clinical Application, CiRA, Kyoto University, Kyoto, Japan
| | - Sou Nakamura
- Department of Clinical Application, CiRA, Kyoto University, Kyoto, Japan
| | - Naoshi Sugimoto
- Department of Clinical Application, CiRA, Kyoto University, Kyoto, Japan
| | - Shunsuke Kihara
- Department of Fundamental Cell Technology, CiRA, Kyoto University, Kyoto, Japan
| | - Masato Nakagawa
- Department of Life Science Frontiers, CiRA, Kyoto University, Kyoto, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, CiRA, Kyoto University, Kyoto, Japan; Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto 606-8501, Japan; AMED-CREST, AMED 1-7-1 Otemachi, Chiyodaku, Tokyo 100-0004, Japan
| | - Kiyotoshi Sekiguchi
- Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Japan
| | - Koji Eto
- Department of Clinical Application, CiRA, Kyoto University, Kyoto, Japan; Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba, Japan.
| |
Collapse
|
10
|
Gerber N, Brunner MAT, Jagannathan V, Leeb T, Gerhards NM, Welle MM, Dettwiler M. Transcriptional Differences between Canine Cutaneous Epitheliotropic Lymphoma and Immune-Mediated Dermatoses. Genes (Basel) 2021; 12:160. [PMID: 33504055 PMCID: PMC7912288 DOI: 10.3390/genes12020160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 11/16/2022] Open
Abstract
Canine cutaneous epitheliotropic T-cell lymphoma (CETL) and immune-mediated T-cell predominant dermatoses (IMD) share several clinical and histopathological features, but differ substantially in prognosis. The discrimination of ambiguous cases may be challenging, as diagnostic tests are limited and may prove equivocal. This study aimed to investigate transcriptional differences between CETL and IMD, as a basis for further research on discriminating diagnostic biomarkers. We performed 100bp single-end sequencing on RNA extracted from formalin-fixed and paraffin-embedded skin biopsies from dogs with CETL and IMD, respectively. DESeq2 was used for principal component analysis (PCA) and differential gene expression analysis. Genes with significantly different expression were analyzed for enriched pathways using two different tools. The expression of selected genes and their proteins was validated by RT-qPCR and immunohistochemistry. PCA demonstrated the distinct gene expression profiles of CETL and IMD. In total, 503 genes were upregulated, while 4986 were downregulated in CETL compared to IMD. RT-qPCR confirmed the sequencing results for 5/6 selected genes tested, while the protein expression detected by immunohistochemistry was not entirely consistent. Our study revealed transcriptional differences between canine CETL and IMD, with similarities to human cutaneous lymphoma. Differentially expressed genes are potential discriminatory markers, but require further validation on larger sample collections.
Collapse
Affiliation(s)
- Nadja Gerber
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3001 Bern, Switzerland; (N.G.); (M.A.T.B.); (N.M.G.); (M.M.W.)
- Dermfocus, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland; (V.J.); (T.L.)
- Grosstierpraxis Weibel + Werner, Oberdorfstrasse 15, 3438 Lauperswil, Switzerland
| | - Magdalena A. T. Brunner
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3001 Bern, Switzerland; (N.G.); (M.A.T.B.); (N.M.G.); (M.M.W.)
- Dermfocus, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland; (V.J.); (T.L.)
| | - Vidhya Jagannathan
- Dermfocus, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland; (V.J.); (T.L.)
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109A, 3001 Bern, Switzerland
| | - Tosso Leeb
- Dermfocus, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland; (V.J.); (T.L.)
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109A, 3001 Bern, Switzerland
| | - Nora M. Gerhards
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3001 Bern, Switzerland; (N.G.); (M.A.T.B.); (N.M.G.); (M.M.W.)
- Wageningen Bioveterinary Research, Wageningen University & Research, Houtribweg 39, 8221 RA Lelystad, The Netherlands
| | - Monika M. Welle
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3001 Bern, Switzerland; (N.G.); (M.A.T.B.); (N.M.G.); (M.M.W.)
- Dermfocus, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland; (V.J.); (T.L.)
| | - Martina Dettwiler
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3001 Bern, Switzerland; (N.G.); (M.A.T.B.); (N.M.G.); (M.M.W.)
- Dermfocus, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland; (V.J.); (T.L.)
| |
Collapse
|
11
|
Integrin-linked kinase controls retinal angiogenesis and is linked to Wnt signaling and exudative vitreoretinopathy. Nat Commun 2019; 10:5243. [PMID: 31748531 PMCID: PMC6868140 DOI: 10.1038/s41467-019-13220-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 10/18/2019] [Indexed: 01/26/2023] Open
Abstract
Familial exudative vitreoretinopathy (FEVR) is a human disease characterized by defective retinal angiogenesis and associated complications that can result in vision loss. Defective Wnt/β-catenin signaling is an established cause of FEVR, whereas other molecular alterations contributing to the disease remain insufficiently understood. Here, we show that integrin-linked kinase (ILK), a mediator of cell-matrix interactions, is indispensable for retinal angiogenesis. Inactivation of the murine Ilk gene in postnatal endothelial cells results in sprouting defects, reduced endothelial proliferation and disruption of the blood-retina barrier, resembling phenotypes seen in established mouse models of FEVR. Retinal vascularization defects are phenocopied by inducible inactivation of the gene for α-parvin (Parva), an interactor of ILK. Screening genomic DNA samples from exudative vitreoretinopathy patients identifies three distinct mutations in human ILK, which compromise the function of the gene product in vitro. Together, our data suggest that defective cell-matrix interactions are linked to Wnt signaling and FEVR. Integrin-linked kinase (ILK) is an important mediator of integrin signaling. Here Park et al. show that mice with endothelial-specific deletion of Ilk develop vascular defects that resemble familial exudative vitreoretinopathy, and identify mutations in ILK in patients with exudative vitreoretinopathy suggesting a potential role in human pathogenesis.
Collapse
|
12
|
Kelaini S, Vilà-González M, Caines R, Campbell D, Eleftheriadou M, Tsifaki M, Magee C, Cochrane A, O'neill K, Yang C, Stitt AW, Zeng L, Grieve DJ, Margariti A. Follistatin-Like 3 Enhances the Function of Endothelial Cells Derived from Pluripotent Stem Cells by Facilitating β-Catenin Nuclear Translocation Through Inhibition of Glycogen Synthase Kinase-3β Activity. Stem Cells 2018; 36:1033-1044. [PMID: 29569797 PMCID: PMC6099345 DOI: 10.1002/stem.2820] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 02/10/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Abstract
The fight against vascular disease requires functional endothelial cells (ECs) which could be provided by differentiation of induced Pluripotent Stem Cells (iPS Cells) in great numbers for use in the clinic. However, the great promise of the generated ECs (iPS-ECs) in therapy is often restricted due to the challenge in iPS-ECs preserving their phenotype and function. We identified that Follistatin-Like 3 (FSTL3) is highly expressed in iPS-ECs, and, as such, we sought to clarify its possible role in retaining and improving iPS-ECs function and phenotype, which are crucial in increasing the cells' potential as a therapeutic tool. We overexpressed FSTL3 in iPS-ECs and found that FSTL3 could induce and enhance endothelial features by facilitating β-catenin nuclear translocation through inhibition of glycogen synthase kinase-3β activity and induction of Endothelin-1. The angiogenic potential of FSTL3 was also confirmed both in vitro and in vivo. When iPS-ECs overexpressing FSTL3 were subcutaneously injected in in vivo angiogenic model or intramuscularly injected in a hind limb ischemia NOD.CB17-Prkdcscid/NcrCrl SCID mice model, FSTL3 significantly induced angiogenesis and blood flow recovery, respectively. This study, for the first time, demonstrates that FSTL3 can greatly enhance the function and maturity of iPS-ECs. It advances our understanding of iPS-ECs and identifies a novel pathway that can be applied in cell therapy. These findings could therefore help improve efficiency and generation of therapeutically relevant numbers of ECs for use in patient-specific cell-based therapies. In addition, it can be particularly useful toward the treatment of vascular diseases instigated by EC dysfunction. Stem Cells 2018;36:1033-1044.
Collapse
Affiliation(s)
- Sophia Kelaini
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Marta Vilà-González
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Rachel Caines
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - David Campbell
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | | | - Marianna Tsifaki
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Corey Magee
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Amy Cochrane
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Karla O'neill
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Chunbo Yang
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Alan W Stitt
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Lingfang Zeng
- Cardiovascular Division, King's College London, London, United Kingdom
| | - David J Grieve
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Andriana Margariti
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
13
|
Matias D, Dubois LG, Pontes B, Rosário L, Ferrer VP, Balça-Silva J, Fonseca ACC, Macharia LW, Romão L, E Spohr TCLDS, Chimelli L, Filho PN, Lopes MC, Abreu JG, Lima FRS, Moura-Neto V. GBM-Derived Wnt3a Induces M2-Like Phenotype in Microglial Cells Through Wnt/β-Catenin Signaling. Mol Neurobiol 2018; 56:1517-1530. [PMID: 29948952 DOI: 10.1007/s12035-018-1150-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 05/23/2018] [Indexed: 12/14/2022]
Abstract
Glioblastoma is an extremely aggressive and deadly brain tumor known for its striking cellular heterogeneity and capability to communicate with microenvironment components, such as microglia. Microglia-glioblastoma interaction contributes to an increase in tumor invasiveness, and Wnt signaling pathway is one of the main cascades related to tumor progression through changes in cell migration and invasion. However, very little is known about the role of canonical Wnt signaling during microglia-glioblastoma crosstalk. Here, we show for the first time that Wnt3a is one of the factors that regulate interactions between microglia and glioblastoma cells. Wnt3a activates the Wnt/β-catenin signaling of both glioblastoma and microglial cells. Glioblastoma-conditioned medium not only induces nuclear translocation of microglial β-catenin but also increases microglia viability and proliferation as well as Wnt3a, cyclin-D1, and c-myc expression. Moreover, glioblastoma-derived Wnt3a increases microglial ARG-1 and STI1 expression, followed by an upregulation of IL-10 mRNA levels, and a decrease in IL1β gene expression. The presence of Wnt3a in microglia-glioblastoma co-cultures increases the formation of membrane nanotubes accompanied by changes in migration capability. In vivo, tumors formed from Wnt3a-stimulated glioblastoma cells presented greater microglial infiltration and more aggressive characteristics such as growth rate than untreated tumors. Thus, we propose that Wnt3a belongs to the arsenal of factors capable of stimulating the induction of M2-like phenotype on microglial cells, which contributes to the poor prognostic of glioblastoma, reinforcing that Wnt/β-catenin pathway can be a potential therapeutic target to attenuate glioblastoma progression.
Collapse
Affiliation(s)
- Diana Matias
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Saúde do Estado do Rio de Janeiro, Rua do Resende 156, Rio de Janeiro, CEP 20231-092, Brazil.,Instituto de Ciências Biomédicas da Universidade Federal do Rio de Janeiro (ICB/UFRJ), Rio de Janeiro, 21941-902, Brazil
| | - Luiz Gustavo Dubois
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Saúde do Estado do Rio de Janeiro, Rua do Resende 156, Rio de Janeiro, CEP 20231-092, Brazil.,Instituto de Ciências Biomédicas da Universidade Federal do Rio de Janeiro (ICB/UFRJ), Rio de Janeiro, 21941-902, Brazil
| | - Bruno Pontes
- Instituto de Ciências Biomédicas da Universidade Federal do Rio de Janeiro (ICB/UFRJ), Rio de Janeiro, 21941-902, Brazil
| | - Luciane Rosário
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Saúde do Estado do Rio de Janeiro, Rua do Resende 156, Rio de Janeiro, CEP 20231-092, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina da Universidade Federal do Rio de Janeiro -UFRJ, Rio de Janeiro, Brazil
| | - Valeria Pereira Ferrer
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Saúde do Estado do Rio de Janeiro, Rua do Resende 156, Rio de Janeiro, CEP 20231-092, Brazil
| | - Joana Balça-Silva
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Saúde do Estado do Rio de Janeiro, Rua do Resende 156, Rio de Janeiro, CEP 20231-092, Brazil.,Centro de Neurociências e Biologia celular e Instituto Biomédico da Imagem e das Ciências da Vida (CNC.IBILI), Coimbra, Portugal.,Faculdade de Medicina da Universidade de Coimbra (FMUC), Coimbra, Portugal
| | - Anna Carolina Carvalho Fonseca
- Instituto de Ciências Biomédicas da Universidade Federal do Rio de Janeiro (ICB/UFRJ), Rio de Janeiro, 21941-902, Brazil
| | - Lucy Wanjiku Macharia
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Saúde do Estado do Rio de Janeiro, Rua do Resende 156, Rio de Janeiro, CEP 20231-092, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina da Universidade Federal do Rio de Janeiro -UFRJ, Rio de Janeiro, Brazil
| | - Luciana Romão
- Instituto de Ciências Biomédicas da Universidade Federal do Rio de Janeiro (ICB/UFRJ), Rio de Janeiro, 21941-902, Brazil.,Campus Duque de Caxias, Universidade Federal do Rio de Janeiro, Duque de Caxias, Brazil
| | - Tania Cristina Leite de Sampaio E Spohr
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Saúde do Estado do Rio de Janeiro, Rua do Resende 156, Rio de Janeiro, CEP 20231-092, Brazil
| | - Leila Chimelli
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Saúde do Estado do Rio de Janeiro, Rua do Resende 156, Rio de Janeiro, CEP 20231-092, Brazil
| | - Paulo Niemeyer Filho
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Saúde do Estado do Rio de Janeiro, Rua do Resende 156, Rio de Janeiro, CEP 20231-092, Brazil
| | - Maria Celeste Lopes
- Centro de Neurociências e Biologia celular e Instituto Biomédico da Imagem e das Ciências da Vida (CNC.IBILI), Coimbra, Portugal.,Pólo das Ciências da Saúde, Faculdade de Farmácia da Universidade de Coimbra, Coimbra, Portugal
| | - José Garcia Abreu
- Instituto de Ciências Biomédicas da Universidade Federal do Rio de Janeiro (ICB/UFRJ), Rio de Janeiro, 21941-902, Brazil
| | - Flavia Regina Souza Lima
- Instituto de Ciências Biomédicas da Universidade Federal do Rio de Janeiro (ICB/UFRJ), Rio de Janeiro, 21941-902, Brazil
| | - Vivaldo Moura-Neto
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Saúde do Estado do Rio de Janeiro, Rua do Resende 156, Rio de Janeiro, CEP 20231-092, Brazil.
| |
Collapse
|
14
|
Han S, Pang MF, Nelson CM. Substratum stiffness tunes proliferation downstream of Wnt3a in part by regulating integrin-linked kinase and frizzled-1. J Cell Sci 2018; 131:jcs.210476. [PMID: 29588395 DOI: 10.1242/jcs.210476] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 02/27/2018] [Indexed: 12/14/2022] Open
Abstract
The Wnt/β-catenin pathway controls a variety of cellular behaviors, aberrant activation of which are associated with tumor progression in several types of cancer. The same cellular behaviors are also affected by the mechanical properties of the extracellular matrix (ECM) substratum, which induces signaling through integrins and integrin-linked kinase (ILK). Here, we examined the role of substratum stiffness in the regulation of cell proliferation downstream of Wnt3a. We found that treatment with Wnt3a increased proliferation of cells cultured on stiff substrata, with compliances characteristic of breast tumors, but not of cells on soft substrata, with compliances comparable to that of normal mammary tissue. Depleting ILK rendered cells unresponsive to Wnt3a on both substrata. Ectopic expression of ILK permitted Wnt3a to induce proliferation of cells on both microenvironments, although proliferation on soft substrata remained lower than that on stiff substrata. We further showed that ILK regulates expression of the Wnt receptor frizzled-1 (Fzd1), suggesting the presence of a positive feedback loop between Wnt3a, ILK and Fzd1. These findings suggest that tissue mechanics regulates the cellular response to Wnt under physiological and pathological microenvironmental conditions.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Siyang Han
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Mei-Fong Pang
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Celeste M Nelson
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA .,Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
15
|
Ren C, Chen X, Du N, Geng S, Hu Y, Liu X, Wu X, Lin Y, Bai X, Yin W, Cheng S, Yang L, Zhang Y. Low-intensity pulsed ultrasound promotes Schwann cell viability and proliferation via the GSK-3β/β-catenin signaling pathway. Int J Biol Sci 2018; 14:497-507. [PMID: 29805301 PMCID: PMC5968842 DOI: 10.7150/ijbs.22409] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 02/16/2018] [Indexed: 12/17/2022] Open
Abstract
Background: It has been reported that ultrasound enhances peripheral nerve regeneration, but the mechanism remains elusive. Low-intensity pulsed ultrasound (LIPUS) has been reported to enhance proliferation and alter protein production in various types of cells. In this study, we detected the effects of LIPUS on Schwann cells. Material and methods: Schwann cells were separated from new natal Sprague-Dawley rat sciatic nerves and were cultured and purified. The Schwann cells were treated by LIPUS for 10 minutes every day, with an intensity of 27.37 mW/cm2. After treatment for 5 days, MTT, EdU staining, and flow cytometry were performed to examine cell viability and proliferation. Neurotrophic factors, including FGF, NGF, BDNF, and GDNF, were measured by western blot and real-time PCR. GSK-3β, p-GSK-3β, β-catenin and Cyclin D1 protein levels were detected using a western blot analysis. The expression of Cyclin D1 was also detected by immunofluorescence. Results: MTT and EdU staining showed that LIPUS increased the Schwann cells viability and proliferation. Compared to the control group, LIPUS increased the expression of growth factors and neurotrophic factors, including FGF, NGF, BDNF, GDNF, and Cyclin D1. Meanwhile, GSK-3β activity was inhibited in the LIPUS group as demonstrated by the increased level of p-GSK-3β and the ratio of the p-GSK-3β/GSK-3β level. The mRNA and protein expressions of β-catenin were increased in the LIPUS group. However, SB216763, a GSK-3β inhibitor, reversed the effects of LIPUS on Schwann cells. Conclusion: LIPUS promotes Schwann cell viability and proliferation by increasing Cyclin D1 expression via enhancing the GSK-3β/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Cong Ren
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150086, China
| | - Xiaohui Chen
- Departmentof Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, eilongjiang Province 150081, China
| | - Ning Du
- Departmentof Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, eilongjiang Province 150081, China
| | - Shuo Geng
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, China
| | - Yingying Hu
- Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xin Liu
- Departmentof Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, eilongjiang Province 150081, China
| | - Xianxian Wu
- Departmentof Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, eilongjiang Province 150081, China
| | - Yuan Lin
- Departmentof Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, eilongjiang Province 150081, China
| | - Xue Bai
- Departmentof Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, eilongjiang Province 150081, China
| | - Wenzhe Yin
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150086, China
| | - Shi Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150086, China
| | - Lei Yang
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, China
| | - Yong Zhang
- Departmentof Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin, eilongjiang Province 150081, China
- Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin, Heilongjiang Province 150086, China
| |
Collapse
|
16
|
Lin YC, Liu LC, Ho CT, Hung CM, Way TD. Luteolin inhibits ER-α expression through ILK inhibition is regulated by a pathway involving Twist and YB-1. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.12.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
17
|
Dejaeger M, Böhm AM, Dirckx N, Devriese J, Nefyodova E, Cardoen R, St-Arnaud R, Tournoy J, Luyten FP, Maes C. Integrin-Linked Kinase Regulates Bone Formation by Controlling Cytoskeletal Organization and Modulating BMP and Wnt Signaling in Osteoprogenitors. J Bone Miner Res 2017; 32:2087-2102. [PMID: 28574598 DOI: 10.1002/jbmr.3190] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 05/28/2017] [Accepted: 05/31/2017] [Indexed: 12/19/2022]
Abstract
Cell-matrix interactions constitute a fundamental aspect of skeletal cell biology and play essential roles in bone homeostasis. These interactions are primarily mediated by transmembrane integrin receptors, which mediate cell adhesion and transduce signals from the extracellular matrix to intracellular responses via various downstream effectors, including integrin-linked kinase (ILK). ILK functions as adaptor protein at focal adhesion sites, linking integrins to the actin cytoskeleton, and has been reported to act as a kinase phosphorylating signaling molecules such as GSK-3β and Akt. Thereby, ILK plays important roles in cellular attachment, motility, proliferation and survival. To assess the in vivo role of ILK signaling in osteoprogenitors and the osteoblast lineage cells descending thereof, we generated conditional knockout mice using the Osx-Cre:GFP driver strain. Mice lacking functional ILK in osterix-expressing cells and their derivatives showed no apparent developmental or growth phenotype, but by 5 weeks of age they displayed a significantly reduced trabecular bone mass, which persisted into adulthood in male mice. Histomorphometry and serum analysis indicated no alterations in osteoclast formation and activity, but provided evidence that osteoblast function was impaired, resulting in reduced bone mineralization and increased accumulation of unmineralized osteoid. In vitro analyses further substantiated that absence of ILK in osteogenic cells was associated with compromised collagen matrix production and mineralization. Mechanistically, we found evidence for both impaired cytoskeletal functioning and reduced signal transduction in osteoblasts lacking ILK. Indeed, loss of ILK in primary osteogenic cells impaired F-actin organization, cellular adhesion, spreading, and migration, indicative of defective coupling of cell-matrix interactions to the cytoskeleton. In addition, BMP/Smad and Wnt/β-catenin signaling was reduced in the absence of ILK. Taken together, these data demonstrate the importance of integrin-mediated cell-matrix interactions and ILK signaling in osteoprogenitors in the control of osteoblast functioning during juvenile bone mass acquisition and adult bone remodeling and homeostasis. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Marian Dejaeger
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Anna-Marei Böhm
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Naomi Dirckx
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Joke Devriese
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Elena Nefyodova
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Ruben Cardoen
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - René St-Arnaud
- Shriners Hospital for Children, McGill University, Montreal, Canada
| | - Jos Tournoy
- Geriatric Medicine, University Hospitals Leuven, Leuven, Belgium.,Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Frank P Luyten
- Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Christa Maes
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
18
|
Yuan D, Zhao Y, Wang Y, Che J, Tan W, Jin Y, Wang F, Li P, Fu S, Liu Q, Zhu W. Effect of integrin‑linked kinase gene silencing on microRNA expression in ovarian cancer. Mol Med Rep 2017; 16:7267-7276. [PMID: 28944870 PMCID: PMC5865855 DOI: 10.3892/mmr.2017.7523] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 07/11/2017] [Indexed: 12/15/2022] Open
Abstract
Integrin‑linked kinase (ILK) is overexpressed in ovarian cancer (OC), and ILK gene silencing results in apoptosis in OC cells. In the present study, the mechanism by which ILK induces apoptosis was explored from the perspective of microRNA (miRNA) expression. Alterations in the global miRNA expression profile were detected using a miRNA microarray after OC cells were transduced with an ILK small hairpin RNA lentivirus. ILK silencing led to a significant upregulation of 14 miRNAs by at least 1.5‑fold. These findings were validated by reverse transcription‑quantitative polymerase chain reaction. A pathway analysis of experimentally validated target genes revealed the inhibition of multiple cancer‑associated signaling pathways and the wnt signaling pathway. Compared with cells transfected with scrambled RNA, the ILK‑silenced cells had remarkably lower expression of wnt ligands (wnt3a, wnt4 and wnt5a) and downstream β‑catenin. ILK silencing led to apoptosis of OC cells and impaired the migratory ability. Taken together, the present results suggested that miRNA‑mediated wnt pathway alterations are involved in the anti‑apoptotic role of ILK in OC. It was also indicated that ILK silencing reduced the ability of OC cells to adhere to fibronectin, which may lead to unstable focal contact. Consistently, the phosphorylation levels of focal adhesion kinase and RAC‑α serine/threonine protein kinase were downregulated. The present work demonstrated the first global miRNA expression profile of OC cells when ILK was inhibited, and this expression profile may provide a basis for the development of biomarkers and therapeutic targets for OC.
Collapse
Affiliation(s)
- Dandan Yuan
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yilei Zhao
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yang Wang
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Jianhua Che
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Wenhua Tan
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yuxia Jin
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Fei Wang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Peiliang Li
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Shuyan Fu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Qian Liu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Wenliang Zhu
- Institute of Clinical Pharmacology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
19
|
Lai KKY, Kweon SM, Chi F, Hwang E, Kabe Y, Higashiyama R, Qin L, Yan R, Wu RP, Lai K, Fujii N, French S, Xu J, Wang JY, Murali R, Mishra L, Lee JS, Ntambi JM, Tsukamoto H. Stearoyl-CoA Desaturase Promotes Liver Fibrosis and Tumor Development in Mice via a Wnt Positive-Signaling Loop by Stabilization of Low-Density Lipoprotein-Receptor-Related Proteins 5 and 6. Gastroenterology 2017; 152:1477-1491. [PMID: 28143772 PMCID: PMC5406249 DOI: 10.1053/j.gastro.2017.01.021] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 01/09/2017] [Accepted: 01/17/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Stearoyl-CoA desaturase (SCD) synthesizes monounsaturated fatty acids (MUFAs) and has been associated with the development of metabolic syndrome, tumorigenesis, and stem cell characteristics. We investigated whether and how SCD promotes liver fibrosis and tumor development in mice. METHODS Rodent primary hepatic stellate cells (HSCs), mouse liver tumor-initiating stem cell-like cells (TICs), and human hepatocellular carcinoma (HCC) cell lines were exposed to Wnt signaling inhibitors and changes in gene expression patterns were analyzed. We assessed the functions of SCD by pharmacologic and conditional genetic manipulation in mice with hepatotoxic or cholestatic induction of liver fibrosis, orthotopic transplants of TICs, or liver tumors induced by administration of diethyl nitrosamine. We performed bioinformatic analyses of SCD expression in HCC vs nontumor liver samples collected from patients, and correlated levels with HCC stage and patient mortality. We performed nano-bead pull-down assays, liquid chromatography-mass spectrometry, computational modeling, and ribonucleoprotein immunoprecipitation analyses to identify MUFA-interacting proteins. We examined the effects of SCD inhibition on Wnt signaling, including the expression and stability of low-density lipoprotein-receptor-related proteins 5 and 6 (LRP5 and LRP6), by immunoblot and quantitative polymerase chain reaction analyses. RESULTS SCD was overexpressed in activated HSC and HCC cells from patients; levels of SCD messenger RNA (mRNA) correlated with HCC stage and patient survival time. In rodent HSCs and TICs, the Wnt effector β-catenin increased sterol regulatory element binding protein 1-dependent transcription of Scd, and β-catenin in return was stabilized by MUFAs generated by SCD. This loop required MUFA inhibition of binding of Ras-related nuclear protein 1 (Ran1) to transportin 1 and reduced nuclear import of elav-like protein 1 (HuR), increasing cytosolic levels of HuR and HuR-mediated stabilization of mRNAs encoding LRP5 and LRP6. Genetic disruption of Scd and pharmacologic inhibitors of SCD reduced HSC activation and TIC self-renewal and attenuated liver fibrosis and tumorigenesis in mice. Conditional disruption of Scd2 in activated HSCs prevented growth of tumors from TICs and reduced the formation of diethyl nitrosamine-induced liver tumors in mice. CONCLUSIONS In rodent HSCs and TICs, we found SCD expression to be regulated by Wnt-β-catenin signaling, and MUFAs produced by SCD provided a forward loop to amplify Wnt signaling via stabilization of Lrp5 and Lrp6 mRNAs, contributing to liver fibrosis and tumor growth. SCD expressed by HSCs promoted liver tumor development in mice. Components of the identified loop linking HSCs and TICs might be therapeutic targets for liver fibrosis and tumors.
Collapse
Affiliation(s)
- Keane K Y Lai
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, University of Southern California, Los Angeles, California
| | - Soo-Mi Kweon
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, University of Southern California, Los Angeles, California
| | - Feng Chi
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, University of Southern California, Los Angeles, California
| | - Edward Hwang
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, University of Southern California, Los Angeles, California
| | - Yasuaki Kabe
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Reiichi Higashiyama
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, University of Southern California, Los Angeles, California
| | - Lan Qin
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, University of Southern California, Los Angeles, California
| | - Rui Yan
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, University of Southern California, Los Angeles, California
| | - Raymond P Wu
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, University of Southern California, Los Angeles, California
| | - Keith Lai
- Department of Anatomic Pathology, Cleveland Clinic, Cleveland, Ohio
| | - Naoaki Fujii
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Samuel French
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, University of Southern California, Los Angeles, California; Harbor-University of California Los Angeles Medical Center, Torrance, California
| | - Jun Xu
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, University of Southern California, Los Angeles, California
| | - Jian-Ying Wang
- Departments of Surgery and Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ramachandran Murali
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, University of Southern California, Los Angeles, California; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Lopa Mishra
- Department of Surgery and Cancer Center, George Washington University, Washington, District of Columbia
| | - Ju-Seog Lee
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James M Ntambi
- Departments of Biochemistry and Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Hidekazu Tsukamoto
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, University of Southern California, Los Angeles, California; Department of Veterans Affairs, Greater Los Angeles Healthcare System, Los Angeles, California.
| |
Collapse
|
20
|
Krishna L, Dhamodaran K, Jayadev C, Chatterjee K, Shetty R, Khora SS, Das D. Nanostructured scaffold as a determinant of stem cell fate. Stem Cell Res Ther 2016; 7:188. [PMID: 28038681 PMCID: PMC5203716 DOI: 10.1186/s13287-016-0440-y] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The functionality of stem cells is tightly regulated by cues from the niche, comprising both intrinsic and extrinsic cell signals. Besides chemical and growth factors, biophysical signals are important components of extrinsic signals that dictate the stem cell properties. The materials used in the fabrication of scaffolds provide the chemical cues whereas the shape of the scaffolds provides the biophysical cues. The effect of the chemical composition of the scaffolds on stem cell fate is well researched. Biophysical signals such as nanotopography, mechanical forces, stiffness of the matrix, and roughness of the biomaterial influence the fate of stem cells. However, not much is known about their role in signaling crosstalk, stem cell maintenance, and directed differentiation. Among the various techniques for scaffold design, nanotechnology has special significance. The role of nanoscale topography in scaffold design for the regulation of stem cell behavior has gained importance in regenerative medicine. Nanotechnology allows manipulation of highly advanced surfaces/scaffolds for optimal regulation of cellular behavior. Techniques such as electrospinning, soft lithography, microfluidics, carbon nanotubes, and nanostructured hydrogel are described in this review, along with their potential usage in regenerative medicine. We have also provided a brief insight into the potential signaling crosstalk that is triggered by nanomaterials that dictate a specific outcome of stem cells. This concise review compiles recent developments in nanoscale architecture and its importance in directing stem cell differentiation for prospective therapeutic applications.
Collapse
Affiliation(s)
- Lekshmi Krishna
- Stem Cell Research Lab, GROW Lab, Narayana Nethralaya Foundation, Bangalore, Karnataka, India.,School of Bioscience and Technology, VIT University, Vellore, Tamil Nadu, India
| | - Kamesh Dhamodaran
- Stem Cell Research Lab, GROW Lab, Narayana Nethralaya Foundation, Bangalore, Karnataka, India.,School of Bioscience and Technology, VIT University, Vellore, Tamil Nadu, India
| | - Chaitra Jayadev
- Vitreoretina Services, Narayana Nethralaya Eye Hospital, Bangalore, Karnataka, India
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science, Bangalore, Karnataka, India
| | - Rohit Shetty
- Department of Cornea and Refractive Surgery, Narayana Nethralaya Eye Hospital, Bangalore, Karnataka, India
| | - S S Khora
- School of Bioscience and Technology, VIT University, Vellore, Tamil Nadu, India
| | - Debashish Das
- Stem Cell Research Lab, GROW Lab, Narayana Nethralaya Foundation, Bangalore, Karnataka, India.
| |
Collapse
|
21
|
Xiong Z, Guo M, Yu Y, Zhang FF, Ge MK, Chen GQ, Shen SM. Downregulation of AIF by HIF-1 contributes to hypoxia-induced epithelial–mesenchymal transition of colon cancer. Carcinogenesis 2016; 37:1079-1088. [DOI: 10.1093/carcin/bgw089] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
22
|
Przybyla L, Muncie JM, Weaver VM. Mechanical Control of Epithelial-to-Mesenchymal Transitions in Development and Cancer. Annu Rev Cell Dev Biol 2016; 32:527-554. [DOI: 10.1146/annurev-cellbio-111315-125150] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Laralynne Przybyla
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, California 94143;
| | - Jonathon M. Muncie
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, California 94143;
- Joint Graduate Group in Bioengineering (University of California, San Francisco, and University of California, Berkeley), San Francisco, California 94143
| | - Valerie M. Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, California 94143;
- Departments of Anatomy, Bioengineering, and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, The Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94143
| |
Collapse
|
23
|
Hsu EC, Kulp SK, Huang HL, Tu HJ, Chao MW, Tseng YC, Yang MC, Salunke SB, Sullivan NJ, Chen WC, Zhang J, Teng CM, Fu WM, Sun D, Wicha MS, Shapiro CL, Chen CS. Integrin-linked kinase as a novel molecular switch of the IL-6-NF-κB signaling loop in breast cancer. Carcinogenesis 2016; 37:430-442. [PMID: 26905583 DOI: 10.1093/carcin/bgw020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 02/05/2016] [Indexed: 12/11/2022] Open
Abstract
Substantial evidence has clearly demonstrated the role of the IL-6-NF-κB signaling loop in promoting aggressive phenotypes in breast cancer. However, the exact mechanism by which this inflammatory loop is regulated remains to be defined. Here, we report that integrin-linked kinase (ILK) acts as a molecular switch for this feedback loop. Specifically, we show that IL-6 induces ILK expression via E2F1 upregulation, which, in turn, activates NF-κB signaling to facilitate IL-6 production. shRNA-mediated knockdown or pharmacological inhibition of ILK disrupted this IL-6-NF-κB signaling loop, and blocked IL-6-induced cancer stem cells in vitro and estrogen-independent tumor growth in vivo Together, these findings establish ILK as an intermediary effector of the IL-6-NF-κB feedback loop and a promising therapeutic target for breast cancer.
Collapse
Affiliation(s)
- En-Chi Hsu
- Division of Medicinal Chemistry and Pharmacognosy , College of Pharmacy , The Ohio State University , Columbus, OH 43210 , USA
| | - Samuel K Kulp
- Division of Medicinal Chemistry and Pharmacognosy , College of Pharmacy , The Ohio State University , Columbus, OH 43210 , USA
| | - Han-Li Huang
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA.,Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Huang-Ju Tu
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA.,Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Min-Wu Chao
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA.,Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Yu-Chou Tseng
- Division of Medicinal Chemistry and Pharmacognosy , College of Pharmacy , The Ohio State University , Columbus, OH 43210 , USA
| | - Ming-Chen Yang
- Division of Medicinal Chemistry and Pharmacognosy , College of Pharmacy , The Ohio State University , Columbus, OH 43210 , USA
| | - Santosh B Salunke
- Division of Medicinal Chemistry and Pharmacognosy , College of Pharmacy , The Ohio State University , Columbus, OH 43210 , USA
| | - Nicholas J Sullivan
- Department of Molecular Virology , Immunology , and Medical Genetics , College of Medicine , The Ohio State University , Columbus , OH 43210 , USA
| | - Wen-Chung Chen
- Department of Pathology , College of Medicine , National Cheng Kung University , Tainan 701 , Taiwan
| | - Jianying Zhang
- Center for Biostatistics , College of Medicine , The Ohio State University , Columbus , OH 43210 , USA
| | - Che-Ming Teng
- Department of Pharmacology , College of Medicine , National Taiwan University , Taipei 10051 , Taiwan
| | - Wen-Mei Fu
- Department of Pharmacology , College of Medicine , National Taiwan University , Taipei 10051 , Taiwan
| | - Duxin Sun
- Department of Pharmaceutical Sciences , College of Pharmacy , University of Michigan , Ann Arbor , MI 48109 , USA
| | - Max S Wicha
- Department of Internal Medicine , University of Michigan Medical School , University of Michigan Comprehensive Cancer Center , Ann Arbor, MI 48109 , USA
| | - Charles L Shapiro
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Mount Sinai Medical Center , New York, NY 10029 , USA and
| | - Ching-Shih Chen
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA.,Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
24
|
MicroRNA-27a-3p Inhibits Melanogenesis in Mouse Skin Melanocytes by Targeting Wnt3a. Int J Mol Sci 2015; 16:10921-33. [PMID: 26006230 PMCID: PMC4463683 DOI: 10.3390/ijms160510921] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 05/06/2015] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) play an essential role in the regulation of almost all the biological processes, including melanogenesis. MiR-27a-3p is nearly six times higher in white alpaca skin compared to brown skin, which indicates that miR-27a-3p may be a candidate regulator for melanogenesis. Wnt3a plays an important role in promoting melanoblasts to differentiate into melanocytes and melanogenesis. To confirm the function of miR-27a-3p to melanogenesis in mammals, miR-27a-3p mimic, inhibitor and their negative control were transfected into mouse melanocytes. As a result, miR-27a-3p inhibits melanogenesis by repressing Wnt3a at post-transcriptional level. A significant decrease in Wnt3a luciferase activity was observed in 293T cells co-transfected with the matched luciferase reporter vector and pre-miR-27a. Furthermore, the presence of exogenous miR-27a-3p significantly decreased Wnt3a protein expression rather than mRNA and reduced β-catenin mRNA levels in melanocytes. The over-expression of miR-27a-3p significantly increased the melanin content of melanocytes. However, miR-27a-3p inhibitor performs an opposite effect on melanogenesis. Wnt3a is one target of miR-27a-3p. MiR-27a-3p could inhibit Wnt3a protein amount by post-transcriptional regulation and melanogenesis in mouse melanocytes. Previous studies reported that Wnt3a promoted melanogenensis in mouse melanocytes. Thus, miR-27-3p inhibits melanogenesis by repressing Wnt3a protein expression.
Collapse
|
25
|
Kalra J, Dragowska WH, Bally MB. Using Pharmacokinetic Profiles and Digital Quantification of Stained Tissue Microarrays as a Medium-Throughput, Quantitative Method for Measuring the Kinetics of Early Signaling Changes Following Integrin-Linked Kinase Inhibition in an In Vivo Model of Cancer. J Histochem Cytochem 2015; 63:691-709. [PMID: 25940338 PMCID: PMC4804727 DOI: 10.1369/0022155415587978] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 04/27/2015] [Indexed: 12/24/2022] Open
Abstract
A small molecule inhibitor (QLT0267) targeting integrin-linked kinase is able to slow breast tumor growth in vivo; however, the mechanism of action remains unknown. Understanding how targeting molecules involved in intersecting signaling pathways impact disease is challenging. To facilitate this understanding, we used tumor tissue microarrays (TMA) and digital image analysis for quantification of immunohistochemistry (IHC) in order to investigate how QLT0267 affects signaling pathways in an orthotopic model of breast cancer over time. Female NCR nude mice were inoculated with luciferase-positive human breast tumor cells (LCC6Luc) and tumor growth was assessed by bioluminescent imaging (BLI). The plasma levels of QLT0267 were determined by LC-MS/MS methods following oral dosing of QLT0267 (200 mg/kg). A TMA was constructed using tumor tissue collected at 2, 4, 6, 24, 78 and 168 hr after treatment. IHC methods were used to assess changes in ILK-related signaling. The TMA was digitized, and Aperio ScanScope and ImageScope software were used to provide semi-quantitative assessments of staining levels. Using medium-throughput IHC quantitation, we show that ILK targeting by QLT0267 in vivo influences tumor physiology through transient changes in pathways involving AKT, GSK-3 and TWIST accompanied by the translocation of the pro-apoptotic protein BAD and an increase in Caspase-3 activity.
Collapse
Affiliation(s)
- Jessica Kalra
- Experimental Therapeutics BC Cancer Agency, British Columbia, Canada (JK,WHD,MBB),Langara College, Vancouver, British Columbia, Canada (JK)
| | - Weislawa H Dragowska
- Experimental Therapeutics BC Cancer Agency, British Columbia, Canada (JK,WHD,MBB)
| | - Marcel B Bally
- Experimental Therapeutics BC Cancer Agency, British Columbia, Canada (JK,WHD,MBB),Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia (MBB),Department of Biochemistry, University of British Columbia, Vancouver, British Columbia (MBB),Faculty of Pharm. Sciences, University of British Columbia, Vancouver, British Columbia (MBB),Center for Drug Research and Development Vancouver, British Columbia, Canada (MBB)
| |
Collapse
|
26
|
Saidak Z, Le Henaff C, Azzi S, Marty C, Da Nascimento S, Sonnet P, Marie PJ. Wnt/β-catenin signaling mediates osteoblast differentiation triggered by peptide-induced α5β1 integrin priming in mesenchymal skeletal cells. J Biol Chem 2015; 290:6903-12. [PMID: 25631051 DOI: 10.1074/jbc.m114.621219] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The α5β1 integrin is a key fibronectin (FN) receptor that binds to RGD-containing peptides to mediate cell adhesion. We previously reported that α5β1 integrin promotes osteogenic differentiation in mesenchymal skeletal cells (MSCs), but the underlying mechanisms are not fully understood. In this study, we determined the signaling mechanisms induced by α5β1 integrin interaction with its high-affinity ligand CRRETAWAC in murine and human MSCs and in vivo. We show that cyclized CRRETAWAC fully displaced MSC adhesion to FN, whereas related peptides lacking the full RRET sequence produced a partial displacement, indicating that RRET acts as an RGD-like sequence that is required to antagonize FN-mediated cell adhesion. However, all peptides increased focal adhesion kinase phosphorylation, OSE2 transcriptional activity, osteoblast gene expression, and matrix mineralization in MSCs, indicating that peptide-induced α5β1 integrin priming can promote osteogenic differentiation independently of the RRET sequence. Biochemical analyses showed that peptide-induced α5β1 integrin priming transiently increased PI3K/Akt phosphorylation and promoted Wnt/β-catenin transcriptional activity independently of RRET. Consistently, pharmacological inhibition of PI3K activity reduced osteoblast differentiation and abolished Wnt regulatory gene expression induced by α5β1 integrin priming. In vivo, systemic delivery of cyclized GACRETAWACGA linked to (DSS)6 to allow delivery to bone-forming sites for 6 weeks increased serum osteocalcin levels and improved long bone mass and microarchitecture in SAMP-6 senescent osteopenic mice. The results support a mechanism whereby α5β1 integrin priming by high-affinity ligands integrates Wnt/β-catenin signaling to promote osteoblast differentiation independently of cell adhesion, which could be used to improve bone mass and microarchitecture in the aging skeleton.
Collapse
Affiliation(s)
- Zuzana Saidak
- From UMR-1132 INSERM, 75475 Paris Cedex 10, France, the Université Paris Diderot, Sorbonne Paris Cité, Paris, France, and
| | - Carole Le Henaff
- From UMR-1132 INSERM, 75475 Paris Cedex 10, France, the Université Paris Diderot, Sorbonne Paris Cité, Paris, France, and
| | - Sofia Azzi
- From UMR-1132 INSERM, 75475 Paris Cedex 10, France, the Université Paris Diderot, Sorbonne Paris Cité, Paris, France, and
| | - Caroline Marty
- From UMR-1132 INSERM, 75475 Paris Cedex 10, France, the Université Paris Diderot, Sorbonne Paris Cité, Paris, France, and
| | - Sophie Da Nascimento
- the Equipe Théra, Laboratoire de Glycochimie, des Antimicrobiens, et des Agroressources (LG2A)-FRE-CNRS 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 80037 Amiens Cedex 1, France
| | - Pascal Sonnet
- the Equipe Théra, Laboratoire de Glycochimie, des Antimicrobiens, et des Agroressources (LG2A)-FRE-CNRS 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 80037 Amiens Cedex 1, France
| | - Pierre J Marie
- From UMR-1132 INSERM, 75475 Paris Cedex 10, France, the Université Paris Diderot, Sorbonne Paris Cité, Paris, France, and
| |
Collapse
|
27
|
Alapati D, Rong M, Chen S, Hehre D, Hummler SC, Wu S. Inhibition of β-catenin signaling improves alveolarization and reduces pulmonary hypertension in experimental bronchopulmonary dysplasia. Am J Respir Cell Mol Biol 2014; 51:104-13. [PMID: 24484510 DOI: 10.1165/rcmb.2013-0346oc] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common and serious chronic lung disease of preterm infants. The development of pulmonary hypertension (PH) significantly increases the mortality and morbidity of this disease. β-Catenin signaling plays an important role in tissue development and remodeling. Aberrant β-catenin signaling is associated with clinical and experiment models of BPD. To test the hypothesis that inhibition of β-catenin signaling is beneficial in promoting alveolar and vascular development and preventing PH in experimental BPD, we examined the effects of ICG001, a newly developed pharmacological inhibitor of β-catenin, in preventing hyperoxia-induced BPD in neonatal rats. Newborn rat pups were randomized at postnatal day (P)2 to room air (RA) + DMSO (placebo), RA + ICG001, 90% FiO2 (O2) + DMSO, or O2 + ICG001. ICG001 (10 mg/kg) or DMSO was given by daily intraperitoneal injection for 14 days during continuous exposure to RA or hyperoxia. Primary human pulmonary arterial smooth muscle cells (PASMCs) were cultured in RA or hyperoxia (95% O2) in the presence of DMSO or ICG001 for 24 to 72 hours. Treatment with ICG001 significantly increased alveolarization and reduced pulmonary vascular remodeling and PH during hyperoxia. Furthermore, administering ICG001 decreased PASMC proliferation and expression of extracellular matrix remodeling molecules in vitro under hyperoxia. Finally, these structural, cellular, and molecular effects of ICG001 were associated with down-regulation of multiple β-catenin target genes. These data indicate that β-catenin signaling mediates hyperoxia-induced alveolar impairment and PH in neonatal animals. Targeting β-catenin may provide a novel strategy to alleviate BPD in preterm infants.
Collapse
Affiliation(s)
- Deepthi Alapati
- Department of Pediatrics, Division of Neonatology, Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | | | | | | | | | | |
Collapse
|
28
|
Ahmed AU, Sarvestani ST, Gantier MP, Williams BRG, Hannigan GE. Integrin-linked kinase modulates lipopolysaccharide- and Helicobacter pylori-induced nuclear factor κB-activated tumor necrosis factor-α production via regulation of p65 serine 536 phosphorylation. J Biol Chem 2014; 289:27776-93. [PMID: 25100717 DOI: 10.1074/jbc.m114.574541] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Integrin-linked kinase (ILK) is a ubiquitously expressed and highly conserved serine-threonine protein kinase that regulates cellular responses to a wide variety of extracellular stimuli. ILK is involved in cell-matrix interactions, cytoskeletal organization, and cell signaling. ILK signaling has also been implicated in oncogenesis and progression of cancers. However, its role in the innate immune system remains unknown. Here, we show that ILK mediates pro-inflammatory signaling in response to lipopolysaccharide (LPS). Pharmacological or genetic inhibition of ILK in mouse embryonic fibroblasts and macrophages selectively blocks LPS-induced production of the pro-inflammatory cytokine tumor necrosis factor α (TNF-α). ILK is required for LPS-induced activation of nuclear factor κB (NF-κB) and transcriptional induction of TNF-α. The modulation of LPS-induced TNF-α synthesis by ILK does not involve the classical NF-κB pathway, because IκB-α degradation and p65 nuclear translocation are both unaffected by ILK inhibition. Instead, ILK is involved in an alternative activation of NF-κB signaling by modulating the phosphorylation of p65 at Ser-536. Furthermore, ILK-mediated alternative NF-κB activation through p65 Ser-536 phosphorylation also occurs during Helicobacter pylori infection in macrophages and gastric cancer cells. Moreover, ILK is required for H. pylori-induced TNF-α secretion in macrophages. Although ILK-mediated phosphorylation of p65 at Ser-536 is independent of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway during LPS stimulation, upon H. pylori infection this event is dependent on the PI3K/Akt pathway. Our findings implicate ILK as a critical regulatory molecule for the NF-κB-mediated pro-inflammatory signaling pathway, which is essential for innate immune responses against pathogenic microorganisms.
Collapse
Affiliation(s)
- Afsar U Ahmed
- From the Centre for Cancer Research, MIMR-PHI Institute of Medical Research, and Department of Molecular and Translational Science, Monash University, Clayton, Victoria 3168, Australia
| | - Soroush T Sarvestani
- From the Centre for Cancer Research, MIMR-PHI Institute of Medical Research, and Department of Molecular and Translational Science, Monash University, Clayton, Victoria 3168, Australia
| | - Michael P Gantier
- From the Centre for Cancer Research, MIMR-PHI Institute of Medical Research, and Department of Molecular and Translational Science, Monash University, Clayton, Victoria 3168, Australia
| | - Bryan R G Williams
- From the Centre for Cancer Research, MIMR-PHI Institute of Medical Research, and Department of Molecular and Translational Science, Monash University, Clayton, Victoria 3168, Australia
| | - Gregory E Hannigan
- From the Centre for Cancer Research, MIMR-PHI Institute of Medical Research, and Department of Molecular and Translational Science, Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
29
|
Li Y, Zhang J, Yan H. Integrin-linked kinase inhibition attenuates permeability of the streptozotocin-induced diabetic rat retina. Cell Biochem Biophys 2014; 67:1467-72. [PMID: 23712867 DOI: 10.1007/s12013-013-9647-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Integrin-linked kinase (ILK), as a multi-functional regulator, has been associated with diabetic retinopathy (DR). In this study, we investigated whether inhibition of ILK could result in therapeutic effects. Diabetes mellitus's rats were induced by streptozotocin (STZ) injection. After 1 weeks induction, rats were injected intraperitoneally daily with ILK inhibitor, QLT0267, at 5 mg/kg. Then, the rats were examined by 4, 8, and 12 weeks after first STZ injection. We found that QLT0267 treatment could not only lower ILK level in retina at as early as 3 weeks after the onset of diabetes but also attenuate retina permeability, which was measured by Evan's blue. Maximum effect was found in 11 weeks treatment. Meanwhile, QLT0267 did not disturbed blood glucose concentration. Furthermore, QLT0267 inhibited Akt (Ser473) activation and reduced expression of HIF1α and VEGF which were evaluated by western blot, real time PCR, and immunohistochemistry. We conclude that ILK may be a new target for DR.
Collapse
Affiliation(s)
- Yangjun Li
- Department of Ophthalmology, Tangdu Hospital, The Fourth Military Medical University of PLA, Xian, People's Republic of China,
| | | | | |
Collapse
|
30
|
Inactivation of the Hippo tumour suppressor pathway by integrin-linked kinase. Nat Commun 2014; 4:2976. [PMID: 24356468 PMCID: PMC3905719 DOI: 10.1038/ncomms3976] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 11/21/2013] [Indexed: 12/18/2022] Open
Abstract
One of the hallmarks of cancers is the silencing of tumour suppressor genes and pathways. The Hippo tumour suppressor pathway is inactivated in many types of cancers, leading to tumour progression and metastasis. However, the mechanisms of pathway inactivation in tumours remain unclear. Here we demonstrate that integrin-linked kinase (ILK) plays a critical role in the suppression of the Hippo pathway via phospho-inhibition of MYPT1-PP1, leading to inactivation of Merlin. Inhibition of ILK in breast, prostate and colon tumour cells results in the activation of the Hippo pathway components MST1 and LATS1 with concomitant inactivation of YAP/TAZ (Yes-associated protein/transcriptional co-activator with PDZ-binding motif) transcriptional co-activators and TEAD-mediated transcription. Genetic deletion of ILK suppresses ErbB2-driven YAP/TAZ activation in mammary tumours, and its pharmacological inhibition suppresses YAP activation and tumour growth in vivo. Our data demonstrate a role for ILK as a multiple receptor proximal regulator of Hippo tumour suppressor pathway and as a cancer therapeutic target. The Hippo tumour suppressor pathway is inactivated in many cancer types, but how this occurs is unclear. Here, the authors show that integrin-linked kinase (ILK) has a role in inhibiting the Hippo pathway and pharmacological inhibition of ILK reduces the size of tumours in mice.
Collapse
|
31
|
Wu X, Wang J, Jiang H, Hu Q, Chen J, Zhang J, Zhu R, Liu W, Li B. Wnt3a activates β1-integrin and regulates migration and adhesion of vascular smooth muscle cells. Mol Med Rep 2014; 9:1159-64. [PMID: 24535659 DOI: 10.3892/mmr.2014.1937] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Accepted: 01/27/2014] [Indexed: 11/06/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) are known to undergo functional changes that contribute to the pathogenesis of atherosclerosis and restenosis. Wnts are a family of secreted glycoproteins that bind to transmembrane Frizzled receptors and initiate signaling cascades with indispensable roles during cell migration, adhesion, proliferation, and survival. The present study reports that wingless-type MMTV integration site family, member 3a (Wnt3a) activates the canonical Wnt pathway in rat VSMCs by triggering the phosphorylation of β-catenin at position Ser675 and GSK-3β at position Ser9. Phosphorylation of these two proteins increases VSMC migration and adhesion. In a search for the downstream mediators of Wnt3a's effects on VSMC migration and adhesion, Wnt3a treatment was observed to increase integrin-linked kinase (ILK) protein expression. ILK is a serine/threonine protein kinase that is thought to control cell migration and adhesion by regulating the affinity of β1-integrin for the extracellular matrix. Wnt3a treatment of VSMCs also activated β1-integrin without changing the quantity of protein expressed on the cell surface. These results demonstrate that Wnt3a enhances migration and adhesion of VSMCs by activating β1-integrin.
Collapse
Affiliation(s)
- Xiaolin Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jichun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qi Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jing Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Rui Zhu
- Department of Cardiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, P.R. China
| | - Wenwei Liu
- Department of Cardiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, P.R. China
| | - Bin Li
- Department of Cardiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, P.R. China
| |
Collapse
|
32
|
Schernthaner M, Leitinger G, Wolinski H, Kohlwein SD, Reisinger B, Barb RA, Graier WF, Heitz J, Groschner K. Enhanced Ca 2+Entry and Tyrosine Phosphorylation Mediate Nanostructure-Induced Endothelial Proliferation. JOURNAL OF NANOMATERIALS 2013; 2013:251063. [PMID: 24729782 PMCID: PMC3982206 DOI: 10.1155/2013/251063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Nanostructured substrates have been recognized to initiate transcriptional programs promoting cell proliferation. Specifically β-catenin has been identified as transcriptional regulator, activated by adhesion to nanostructures. We set out to identify processes responsible for nanostructure-induced endothelial β-catenin signaling. Transmission electron microscopy (TEM) of cell contacts to differently sized polyethylene terephthalate (PET) surface structures (ripples with 250 to 300 nm and walls with 1.5 μm periodicity) revealed different patterns of cell-substrate interactions. Cell adhesion to ripples occurred exclusively on ripple peaks, while cells were attached to walls continuously. The Src kinase inhibitor PP2 was active only in cells grown on ripples, while the Abl inhibitors dasatinib and imatinib suppressed β-catenin translocation on both structures. Moreover, Gd3+ sensitive Ca2+ entry was observed in response to mechanical stimulation or Ca2+ store depletion exclusively in cells grown on ripples. Both PP2 and Gd3+ suppressed β-catenin nuclear translocation along with proliferation in cells grown on ripples but not on walls. Our results suggest that adhesion of endothelial cells to ripple structured PET induces highly specific, interface topology-dependent changes in cellular signalling, characterized by promotion of Gd3+ -sensitive Ca2+ entry and Src/Abl activation. We propose that these signaling events are crucially involved in nanostructure-induced promotion of cell proliferation.
Collapse
Affiliation(s)
| | - Gerd Leitinger
- Department of Cell Biology, Histology and Embryology, Core Facility Ultrastructure Analysis, Center for Medical Research, Medical University Graz, 8010 Graz, Austria
| | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Sepp D. Kohlwein
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Bettina Reisinger
- Institute of Applied Physics, Johannes Kepler University Linz, 4040 Linz, Austria
| | - Ruxandra-A. Barb
- Institute of Applied Physics, Johannes Kepler University Linz, 4040 Linz, Austria
| | - Wolfgang F. Graier
- Institute of Molecular Biology and Biochemistry, Medical University Graz, 8010 Graz, Austria
| | - Johannes Heitz
- Institute of Applied Physics, Johannes Kepler University Linz, 4040 Linz, Austria
| | - Klaus Groschner
- Institute of Biophysics, Medical University Graz, 8010 Graz, Austria
| |
Collapse
|
33
|
The Xenopus Tgfbi is required for embryogenesis through regulation of canonical Wnt signalling. Dev Biol 2013; 379:16-27. [DOI: 10.1016/j.ydbio.2012.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 09/18/2012] [Accepted: 11/12/2012] [Indexed: 10/27/2022]
|
34
|
De Robertis A, Valensin S, Rossi M, Tunici P, Verani M, De Rosa A, Giordano C, Varrone M, Nencini A, Pratelli C, Benicchi T, Bakker A, Hill J, Sangthongpitag K, Pendharkar V, Liu B, Ng FM, Then SW, Jing Tai S, Cheong SM, He X, Caricasole A, Salerno M. Identification and characterization of a small-molecule inhibitor of Wnt signaling in glioblastoma cells. Mol Cancer Ther 2013; 12:1180-9. [PMID: 23619303 DOI: 10.1158/1535-7163.mct-12-1176-t] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common and prognostically unfavorable form of brain tumor. The aggressive and highly invasive phenotype of these tumors makes them among the most anatomically damaging human cancers with a median survival of less than 1 year. Although canonical Wnt pathway activation in cancers has been historically linked to the presence of mutations involving key components of the pathway (APC, β-catenin, or Axin proteins), an increasing number of studies suggest that elevated Wnt signaling in GBM is initiated by several alternative mechanisms that are involved in different steps of the disease. Therefore, inhibition of Wnt signaling may represent a therapeutically relevant approach for GBM treatment. After the selection of a GBM cell model responsive to Wnt inhibition, we set out to develop a screening approach for the identification of compounds capable of modulating canonical Wnt signaling and associated proliferative responses in GBM cells. Here, we show that the small molecule SEN461 inhibits the canonical Wnt signaling pathway in GBM cells, with relevant effects at both molecular and phenotypic levels in vitro and in vivo. These include SEN461-induced Axin stabilization, increased β-catenin phosphorylation/degradation, and inhibition of anchorage-independent growth of human GBM cell lines and patient-derived primary tumor cells in vitro. Moreover, in vivo administration of SEN461 antagonized Wnt signaling in Xenopus embryos and reduced tumor growth in a GBM xenograft model. These data represent the first demonstration that small-molecule-mediated inhibition of Wnt signaling may be a potential approach for GBM therapeutics.
Collapse
Affiliation(s)
- Alessandra De Robertis
- Corresponding Author: Massimiliano Salerno, Siena Biotech Medicine Research Centre, Via del Petriccio e Belriguardo 35, 53100, Siena, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
The WNT signaling pathway from ligand secretion to gene transcription: molecular mechanisms and pharmacological targets. Pharmacol Ther 2013; 138:66-83. [PMID: 23328704 DOI: 10.1016/j.pharmthera.2013.01.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 12/20/2012] [Indexed: 12/22/2022]
Abstract
Wingless/integrase-1 (WNT) signaling is a key pathway regulating various aspects of embryonic development; however it also underlies several pathological conditions in man, including various cancers and fibroproliferative diseases in several organs. Investigating the molecular processes involved in (canonical) WNT signaling will open new avenues for generating new therapeutics to specifically target diseases in which WNT signaling is aberrantly regulated. Here we describe the complexity of WNT signal transduction starting from the processes involved in WNT ligand biogenesis and secretion by WNT producing cells followed by a comprehensive overview of the molecular signaling events ultimately resulting in enhanced transcription of specific genes in WNT receiving cells. Finally, the possible targets for therapeutic intervention and the available pharmacological inhibitors for this complex signaling pathway are discussed.
Collapse
|
36
|
Liang CH, Chiu SY, Hsu IL, Wu YY, Tsai YT, Ke JY, Pan SH, Hsu YC, Li KC, Yang PC, Chen YL, Hong TM. α-Catulin drives metastasis by activating ILK and driving an αvβ3 integrin signaling axis. Cancer Res 2012; 73:428-38. [PMID: 23047866 DOI: 10.1158/0008-5472.can-12-2095] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
α-Catulin is an oncoprotein that helps sustain proliferation by preventing cellular senescence. Here, we report that α-catulin also drives malignant invasion and metastasis. α-Catulin was upregulated in highly invasive non-small cell lung cancer (NSCLC) cell lines, where its ectopic expression or short-hairpin RNA-mediated attenuation enhanced or limited invasion or metastasis, respectively. α-Catulin interacted with integrin-linked kinase (ILK), a serine/threonine protein kinase implicated in cancer cell proliferation, antiapoptosis, invasion, and angiogenesis. Attenuation of ILK or α-catulin reciprocally blocked cell migration and invasion induced by the other protein. Mechanistic investigations revealed that α-catulin activated Akt-NF-κB signaling downstream of ILK, which in turn led to increased expression of fibronectin and integrin αvβ3. Pharmacologic or antibody-mediated blockade of NF-κB or αvβ3 was sufficient to inhibit α-catulin-induced cell migration and invasion. Clinically, high levels of expression of α-catulin and ILK were associated with poor overall survival in patients with NSCLC. Taken together, our study shows that α-catulin plays a critical role in cancer metastasis by activating the ILK-mediated Akt-NF-κB-αvβ3 signaling axis.
Collapse
Affiliation(s)
- Chen-Hsien Liang
- Institute of Basic Medical Sciences, Institute of Oral Medicine, and Graduate Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Epifano C, Perez-Moreno M. Crossroads of integrins and cadherins in epithelia and stroma remodeling. Cell Adh Migr 2012; 6:261-73. [PMID: 22568988 DOI: 10.4161/cam.20253] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Adhesion events mediated by cadherin and integrin adhesion receptors have fundamental roles in the maintenance of the physiological balance of epithelial tissues, and it is well established that perturbations in their normal functional activity and/or changes in their expression are associated with tumorigenesis. Over the last decades, increasing evidence of a dynamic collaborative interaction between these complexes through their shared interactions with cytoskeletal proteins and common signaling pathways has emerged not only as an important regulator of several aspects of epithelial cell behavior, but also as a coordinated adhesion module that senses and transmits signals from and to the epithelia surrounding microenvironment. The tight regulation of their crosstalk is particularly important during epithelial remodeling events that normally take place during morphogenesis and tissue repair, and when defective it leads to cell transformation and aggravated responses of the tumor microenvironment that contribute to tumorigenesis. In this review we highlight some of the interactions that regulate their crosstalk and how this could be implicated in regulating signals across epithelial tissues to sustain homeostasis.
Collapse
Affiliation(s)
- Carolina Epifano
- Epithelial Cell Biology Group, BBVA Foundation-Cancer Cell Biology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | | |
Collapse
|
38
|
Abstract
The liver is the body's most important detoxification organ and has an extreme ability to regenerate. The regeneration process can be divided into three stages: initiation, proliferation and termination. Most of previous studies focus on the initial stage and proliferative stage, while the mechanism for the proper termination of liver regeneration is still poorly understood. The termination stage involves a variety of cytokines and growth factors, which mainly function to inhibit mitogen-mediated liver cell growth-promoting effect and promote the apoptosis of excessively proliferating liver cells. In this paper we will discuss the major factors involved in the termination of liver regeneration.
Collapse
|
39
|
Sun Y, Chen CS, Fu J. Forcing stem cells to behave: a biophysical perspective of the cellular microenvironment. Annu Rev Biophys 2012; 41:519-42. [PMID: 22404680 DOI: 10.1146/annurev-biophys-042910-155306] [Citation(s) in RCA: 297] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Physical factors in the local cellular microenvironment, including cell shape and geometry, matrix mechanics, external mechanical forces, and nanotopographical features of the extracellular matrix, can all have strong influences on regulating stem cell fate. Stem cells sense and respond to these insoluble biophysical signals through integrin-mediated adhesions and the force balance between intracellular cytoskeletal contractility and the resistant forces originated from the extracellular matrix. Importantly, these mechanotransduction processes can couple with many other potent growth-factor-mediated signaling pathways to regulate stem cell fate. Different bioengineering tools and microscale/nanoscale devices have been successfully developed to engineer the physical aspects of the cellular microenvironment for stem cells, and these tools and devices have proven extremely powerful for identifying the extrinsic physical factors and their downstream intracellular signaling pathways that control stem cell functions.
Collapse
Affiliation(s)
- Yubing Sun
- Integrated Biosystems and Biomechanics Laboratory, Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | |
Collapse
|
40
|
Liu N, Liu W, Ma G, Guo KJ, Song SW. Construction and identification of plasmids carrying small interfering RNAs targeting the ILK gene. Shijie Huaren Xiaohua Zazhi 2012; 20:374-379. [DOI: 10.11569/wcjd.v20.i5.374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To construct plasmids carrying small interfering RNAs (siRNAs) targeting the integrin-linked kinase (ILK) gene and assess their effect on ILK expression in pancreatic cancer cells.
METHODS: Three pairs of siRNAs for ILK were designed and used to construct plasmids carrying siRNAs targeting the ILK gene. The recombinant plasmids and negative control plasmids were stably transfected into Panc-1 cells using cationic liposome Lipofectamine. After transfection, ILK mRNA and protein expression was detected by RT-PCR and Western blotting, respectively.
RESULTS: DNA sequencing results indicated that the recombinant plasmids were constructed correctly. After stable transfection of the recombinant plasmids into Panc-1 cells, ILK mRNA and protein expression was significantly inhibited. Transfection of the recombinant plasmid that had the highest knockdown efficiency reduced ILK mRNA and protein expression by 93.01% and 65.69%, respectively. Compared to the non-transfected group and empty plasmid-transfected group, ILK mRNA expression was significantly down-regulated in the experimental group (0.090 ± 0.009 vs 1.147 ± 0.110, 1.005 ± 0.121, both P < 0.01).
CONCLUSION: Three plasmids carrying siRNAs targeting the ILK gene have been constructed successfully and provide a useful tool for studying the function of ILK.
Collapse
|
41
|
Hu Y, Galkin AV, Wu C, Reddy V, Su AI. CAFET algorithm reveals Wnt/PCP signature in lung squamous cell carcinoma. PLoS One 2011; 6:e25807. [PMID: 22016777 PMCID: PMC3189939 DOI: 10.1371/journal.pone.0025807] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 09/12/2011] [Indexed: 11/25/2022] Open
Abstract
We analyzed the gene expression patterns of 138 Non-Small Cell Lung Cancer (NSCLC) samples and developed a new algorithm called Coverage Analysis with Fisher’s Exact Test (CAFET) to identify molecular pathways that are differentially activated in squamous cell carcinoma (SCC) and adenocarcinoma (AC) subtypes. Analysis of the lung cancer samples demonstrated hierarchical clustering according to the histological subtype and revealed a strong enrichment for the Wnt signaling pathway components in the cluster consisting predominantly of SCC samples. The specific gene expression pattern observed correlated with enhanced activation of the Wnt Planar Cell Polarity (PCP) pathway and inhibition of the canonical Wnt signaling branch. Further real time RT-PCR follow-up with additional primary tumor samples and lung cancer cell lines confirmed enrichment of Wnt/PCP pathway associated genes in the SCC subtype. Dysregulation of the canonical Wnt pathway, characterized by increased levels of β-catenin and epigenetic silencing of negative regulators, has been reported in adenocarcinoma of the lung. Our results suggest that SCC and AC utilize different branches of the Wnt pathway during oncogenesis.
Collapse
Affiliation(s)
- Yue Hu
- Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Anna V. Galkin
- Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Chunlei Wu
- Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Venkateshwar Reddy
- Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Andrew I. Su
- Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
42
|
Assi K, Patterson S, Dedhar S, Owen D, Levings M, Salh B. Role of epithelial integrin-linked kinase in promoting intestinal inflammation: effects on CCL2, fibronectin and the T cell repertoire. BMC Immunol 2011; 12:42. [PMID: 21806815 PMCID: PMC3164617 DOI: 10.1186/1471-2172-12-42] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Accepted: 08/01/2011] [Indexed: 12/13/2022] Open
Abstract
Background The role of integrin signaling in mucosal inflammation is presently unknown. Hence, we aimed to investigate the role of epithelial-derived integrin-linked kinase (ILK), a critical integrin signaling intermediary molecule, in colonic inflammation. Methods Conditional intestinal epithelial cell ILK knockout mice were used for assessment of acute and chronic dextran sodium sulfate (DSS) -induced colitis. Disease activity was scored using standard histological scoring, mucosal cytokines were measured using ELISA, chemokines were determined using reverse-transcription polymerase chain reaction, as well as Q-PCR, and intracellular cytokine staining performed using FACS analysis. Results In both acute and chronic DSS-induced colitis, compared to wild-type mice, ILK-ko mice exhibit less weight loss, and have reduced inflammatory scores. In an in vitro model system using HCT116 cells, we demonstrate that si-RNA mediated down-regulation of ILK results in a reduction in monocyte chemoattractant protein 1 (MCP1, CCL2) chemokine expression. A reduction in CCL2 levels is also observed in the tissue lysates of chronically inflamed colons from ILK-ko mice. Examination of mesenteric lymph node lymphocytes from ILK-ko mice reveals that there is a reduction in the levels of IFN gamma using intracellular staining, together with an increase in Foxp3+ T regulatory cells. Immunohistochemistry demonstrates that reduced fibronectin expression characterizes the inflammatory lesions within the colons of ILK-ko mice. Intriguingly, we demonstrate that fibronectin is directly capable of downregulating T regulatory cell development. Conclusions Collectively, the data indicate for the first time that ILK plays a pro-inflammatory role in intestinal inflammation, through effects on chemokine expression, the extracellular matrix and immune tolerance.
Collapse
Affiliation(s)
- Kiran Assi
- Department of Medicine, The Jack Bell Research Centre, 2660 Oak Street, Vancouver, British Columbia, V6H 3Z6, Canada
| | | | | | | | | | | |
Collapse
|
43
|
Watabe H, Furuhama T, Tani-Ishii N, Mikuni-Takagaki Y. Mechanotransduction activates α₅β₁ integrin and PI3K/Akt signaling pathways in mandibular osteoblasts. Exp Cell Res 2011; 317:2642-9. [PMID: 21824471 DOI: 10.1016/j.yexcr.2011.07.015] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 07/14/2011] [Accepted: 07/16/2011] [Indexed: 10/17/2022]
Abstract
It is unclear how bone cells at different sites detect mechanical loading and how site-specific mechanotransduction affects bone homeostasis. To differentiate the anabolic mechanical responses of mandibular cells from those of calvarial and long bone cells, we isolated osteoblasts from C57B6J mouse bones, cultured them for 1week, and subjected them to therapeutic low intensity pulsed ultrasound (LIPUS). While the expression of the marker proteins of osteoblasts and osteocytes such as alkaline phosphatase and FGF23, as well as Wnt1 and β-catenin, was equally upregulated, the expression of mandibular osteoblast messages related to bone remodeling and apoptosis differed from that of messages of other osteoblasts, in that the messages encoding the pro-remodeling protein RANKL and the anti-apoptotic protein Bcl-2 were markedly upregulated from the very low baseline levels. Blockage of the PI3K and α(5)β(1) integrin pathways showed that the mandibular osteoblast required mechanotransduction downstream of α(5)β(1) integrin to upregulate expression of the proteins β-catenin, p-Akt, Bcl-2, and RANKL. Mandibular osteoblasts thus must be mechanically loaded to preserve their capability to promote remodeling and to insure osteoblast survival, both of which maintain intact mandibular bone tissue. In contrast, calvarial Bcl-2 is fully expressed, together with ILK and phosphorylated mTOR, in the absence of LIPUS. The antibody blocking α(5)β(1) integrin suppressed both the baseline expression of all calvarial proteins examined and the LIPUS-induced expression of all mandibular proteins examined. These findings indicate that the cellular environment, in addition to the tridermic origin, determines site-specific bone homeostasis through the remodeling and survival of osteoblastic cells. Differentiated cells of the osteoblastic lineage at different sites transmit signals through transmembrane integrins such as α(5)β(1) integrin in mandibular osteoblasts, whose signaling may play a major role in controlling bone homeostasis.
Collapse
Affiliation(s)
- H Watabe
- Department of Oral Medicine, Kanagawa Dental College, Japan
| | | | | | | |
Collapse
|
44
|
Alapati D, Rong M, Chen S, Hehre D, Rodriguez MM, Lipson KE, Wu S. Connective tissue growth factor antibody therapy attenuates hyperoxia-induced lung injury in neonatal rats. Am J Respir Cell Mol Biol 2011; 45:1169-77. [PMID: 21659659 DOI: 10.1165/rcmb.2011-0023oc] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Despite recent advances in neonatal intensive care and surfactant therapy, bronchopulmonary dysplasia (BPD) continues to be one of the most common long-term pulmonary complications associated with preterm birth. Clinical efforts to prevent and treat BPD have been largely unsuccessful due to its multifactorial nature and poorly understood disease process. Connective tissue growth factor (CTGF) is a matricellular protein that plays an important role in tissue development and remodeling. Previous studies have demonstrated that hyperoxia exposure up-regulates CTGF expression in neonatal rat lungs. Whether CTGF overexpression plays a role in the pathogenesis of BPD, and whether CTGF antagonism has a therapeutic potential for BPD, are unknown. In the present study, we examined CTGF expression in lung autopsy specimens from patients with BPD and control subjects with no BPD. We assessed the effect of a CTGF-neutralizing monoclonal antibody (CTGF Ab) on preventing hyperoxia-induced lung injury in neonatal rats. Our study demonstrates that CTGF expression is increased in BPD lungs. In newborn rats, exposure to 90% oxygen for 14 days resulted in activation of β-catenin signaling, decreased alveolarization and vascular development, and physiological and histological evidence of pulmonary hypertension (PH). However, treatment with CTGF Ab prevented β-catenin signaling activation, improved alveolarization and vascular development, and attenuated PH during hyperoxia. These data indicate that CTGF-β-catenin signaling plays a critical role in the pathogenesis of experimental BPD. CTGF antagonism may offer a novel therapeutic strategy to alleviate BPD and PH in neonates.
Collapse
Affiliation(s)
- Deepthi Alapati
- Department of Pediatrics, Division of Neonatology, Batchelor Children’s Research Institute, Miami, FL, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Bielefeld KA, Amini-Nik S, Whetstone H, Poon R, Youn A, Wang J, Alman BA. Fibronectin and beta-catenin act in a regulatory loop in dermal fibroblasts to modulate cutaneous healing. J Biol Chem 2011; 286:27687-97. [PMID: 21652705 DOI: 10.1074/jbc.m111.261677] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
β-Catenin is an important regulator of dermal fibroblasts during cutaneous wound repair. However, the factors that modulate β-catenin activity in this process are not completely understood. We investigated the role of the extracellular matrix in regulating β-catenin and found an increase in β-catenin-mediated Tcf-dependent transcriptional activity in fibroblasts exposed to various extracellular matrix components. This occurs through an integrin-mediated GSK3β-dependent pathway. The physiologic role of this mechanism was demonstrated during wound repair in extra domain A-fibronectin-deficient mice, which exhibited decreased β-catenin-mediated signaling during the proliferative phase of healing. Extra domain A-fibronectin-deficient mice have wounds that fail at a lower tensile strength and contain fewer fibroblasts compared with wild type mice. This phenotype was rescued by genetic or pharmacologic activation of β-catenin signaling. Because fibronectin is a transcriptional target of β-catenin, this suggests the existence of a feedback loop between these two molecules that regulates dermal fibroblast cell behavior during wound repair.
Collapse
Affiliation(s)
- Kirsten A Bielefeld
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Ontario M5G 1L7, Canada
| | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Integrin-linked kinase (ILK) is a highly evolutionarily conserved intracellular protein that was originally identified as an integrin-interacting protein, and extensive genetic and biochemical studies have shown that ILK expression is vital during both embryonic development and tissue homeostasis. At the cellular and tissue levels, ILK regulates signaling pathways for cell adhesion-mediated cell survival (anoikis), apoptosis, proliferation and mitosis, migration, invasion, and vascularization and tumor angiogenesis. ILK also has central roles in cardiac and smooth-muscle contractility, and ILK dysregulation causes cardiomyopathies in humans. ILK protein levels are increased in several human cancers and often the expression level predicts poor patient outcome. Abundant evidence has accumulated suggesting that, of the diverse functions of ILK, some may require kinase activity whereas others depend on protein-protein interactions and are, therefore, independent of kinase activity. However, the past several years have seen an ongoing debate about whether ILK indeed functions as a protein serine/threonine kinase. This debate centers on the atypical protein kinase domain of ILK, which lacks some amino-acid residues thought to be essential for phosphotransferase activity. However, similar deficiencies are present in the catalytic domains of other kinases now known to possess protein kinase activity. Numerous studies have shown that ILK phosphorylates peptide substrates in vitro, corresponding to ILK-mediated phosphorylations in intact cells, and a recent report characterizing in vitro phosphotransferase activity of highly purified, full-length ILK, accompanied by detailed enzyme kinetic analyses, shows that, at least in vitro, ILK is a bona fide protein kinase. However, several genetic studies suggest that, not all biological functions of ILK require kinase activity, and that it can function as an adaptor/scaffold protein. Here, we review evidence for and against ILK being an active kinase, and provide a framework for strategies to further analyze the kinase and adaptor functions of ILK in different cellular contexts.
Collapse
|
47
|
Fonar Y, Gutkovich YE, Root H, Malyarova A, Aamar E, Golubovskaya VM, Elias S, Elkouby YM, Frank D. Focal adhesion kinase protein regulates Wnt3a gene expression to control cell fate specification in the developing neural plate. Mol Biol Cell 2011; 22:2409-21. [PMID: 21551070 PMCID: PMC3128541 DOI: 10.1091/mbc.e10-12-0932] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
FAK is linked to aggressive tumors, but its normal function is not clear. FAK knockdown early in Xenopus development anteriorizes the embryo via a loss of Wnt signaling. Wnt3a expression is FAK dependent in both embryos and human breast cancer cells, suggesting that a FAK–Wnt linkage is highly conserved. Focal adhesion kinase (FAK) is a cytoplasmic tyrosine kinase protein localized to regions called focal adhesions, which are contact points between cells and the extracellular matrix. FAK protein acts as a scaffold to transfer adhesion-dependent and growth factor signals into the cell. Increased FAK expression is linked to aggressive metastatic and invasive tumors. However, little is known about its normal embryonic function. FAK protein knockdown during early Xenopus laevis development anteriorizes the embryo. Morphant embryos express increased levels of anterior neural markers, with reciprocally reduced posterior neural marker expression. Posterior neural plate folding and convergence-extension is also inhibited. This anteriorized phenotype resembles that of embryos knocked down zygotically for canonical Wnt signaling. FAK and Wnt3a genes are both expressed in the neural plate, and Wnt3a expression is FAK dependent. Ectopic Wnt expression rescues this FAK morphant anteriorized phenotype. Wnt3a thus acts downstream of FAK to balance anterior–posterior cell fate specification in the developing neural plate. Wnt3a gene expression is also FAK dependent in human breast cancer cells, suggesting that this FAK–Wnt linkage is highly conserved. This unique observation connects the FAK- and Wnt-signaling pathways, both of which act to promote cancer when aberrantly activated in mammalian cells.
Collapse
Affiliation(s)
- Yuri Fonar
- Department of Biochemistry, Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Buske P, Galle J, Barker N, Aust G, Clevers H, Loeffler M. A comprehensive model of the spatio-temporal stem cell and tissue organisation in the intestinal crypt. PLoS Comput Biol 2011; 7:e1001045. [PMID: 21253562 PMCID: PMC3017108 DOI: 10.1371/journal.pcbi.1001045] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2010] [Accepted: 12/01/2010] [Indexed: 12/20/2022] Open
Abstract
We introduce a novel dynamic model of stem cell and tissue organisation in murine intestinal crypts. Integrating the molecular, cellular and tissue level of description, this model links a broad spectrum of experimental observations encompassing spatially confined cell proliferation, directed cell migration, multiple cell lineage decisions and clonal competition. Using computational simulations we demonstrate that the model is capable of quantitatively describing and predicting the dynamic behaviour of the intestinal tissue during steady state as well as after cell damage and following selective gain or loss of gene function manipulations affecting Wnt- and Notch-signalling. Our simulation results suggest that reversibility and flexibility of cellular decisions are key elements of robust tissue organisation of the intestine. We predict that the tissue should be able to fully recover after complete elimination of cellular subpopulations including subpopulations deemed to be functional stem cells. This challenges current views of tissue stem cell organisation. In the murine small intestine there are more than a million organized groups of proliferating cells, the crypts, each of which contains about 250–300 cells. About 60% of these cells are in rapid cycle. The functional stem cells of this tissue have been demonstrated to reside at defined positions at the lower third of the crypt and to give rise to four different cell types. Considering this simple structure the murine intestine is an ideal system to study general aspects of tissue organization. Here, we introduce a comprehensive and predictive computer model of the spatio-temporal organization of the murine intestine which describes how cell production and cell fate decisions could be organized in steady state as well as under perturbations. The model is based on single cells acting as individual agents, updating their status within a certain set of options governed by some active rules and on signals received from the environment. This kind of self-organization enables effective tissue regeneration without assuming an explicit stem cell population that maintains itself by asymmetric division. Thus, the model offers a novel systems biological view on crypt stem cell and tissue organisation.
Collapse
Affiliation(s)
- Peter Buske
- Interdisciplinary Centre for Bioinformatics, University Leipzig, Leipzig, Germany.
| | | | | | | | | | | |
Collapse
|
49
|
Cortez V, Nair BC, Chakravarty D, Vadlamudi RK. Integrin-linked kinase 1: role in hormonal cancer progression. Front Biosci (Schol Ed) 2011; 3:788-96. [PMID: 21196412 DOI: 10.2741/s187] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Integrin-linked kinase 1 (ILK1) is a serine/threonine kinase that plays important roles in a variety of cellular functions including cell survival, migration and angiogenesis. ILK1 is normally expressed in numerous tissues and activated by growth factors, cytokines and hormones. Dysregulation of ILK1 expression or function is found in several hormonal tumors including breast, ovary and prostate. Emerging evidence suggests that ILK overexpression promotes cellular transformation, cell survival, epithelial mesenchymal transition (EMT), and metastasis of hormonal cancer cells while inhibition of ILK1 reduces tumor growth and progression. The recent development of ILK1 inhibitors has provided novel mechanisms for blocking ILK1 signaling to curb metastasis and therapy resistance of hormonal tumors. This review will focus on recent advances made towards understanding the role of ILK signaling axis in progression of hormonal cancer.
Collapse
Affiliation(s)
- Valerie Cortez
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | | | | | | |
Collapse
|
50
|
Rallis C, Pinchin SM, Ish-Horowicz D. Cell-autonomous integrin control of Wnt and Notch signalling during somitogenesis. Development 2010; 137:3591-601. [DOI: 10.1242/dev.050070] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Integrins act at signalling crossroads, and their interactions with other signal transduction pathways are key to the regulation of normal and pathological cell cytoarchitecture and behaviour. Here, we describe a signalling cascade that acts during the formation of the defining segmental features of the vertebrate body – the somites – in which β1-integrin activity regulates epithelialisation by controlling downstream Wnt and Notch activity crucial for somite border formation. Using in vivo transcriptional inhibition in the developing chick embryo, we show that β1-integrin in the anterior presomitic mesoderm activates canonical Wnt signalling in a cell-autonomous, `outside-inside' manner. Signalling is mediated by integrin-linked kinase (ILK), leading to modulation of glycogen synthase kinase 3β (GSK3β) phosphorylation, and activates Notch signalling in the anterior presomitic mesoderm. The two signalling pathways then cooperate to promote somite formation via cMESO1/Mesp2. Our results show that β1-integrin can regulate cell shape and tissue morphogenesis indirectly, by regulation of downstream signalling cascades.
Collapse
Affiliation(s)
- Charalampos Rallis
- Developmental Genetics Laboratory, London Research Institute, Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 4LY, UK
| | - Sheena M. Pinchin
- Developmental Genetics Laboratory, London Research Institute, Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 4LY, UK
| | - David Ish-Horowicz
- Developmental Genetics Laboratory, London Research Institute, Cancer Research UK, 44 Lincoln's Inn Fields, London WC2A 4LY, UK
| |
Collapse
|