1
|
The aminopeptidase B (Ap-B) is phosphorylated in HEK293 cells. Biochimie 2022; 201:204-212. [PMID: 35952945 DOI: 10.1016/j.biochi.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/22/2022]
Abstract
Proteolysis is a post-translational modification (PTM) that affects the whole proteome. First regarded as only destructive, it is more precise than expected. It is finely regulated by other PTMs like phosphorylation. Aminopeptidase B (Ap-B), a M1 metallopeptidase, hydrolyses the peptide bond on the carbonyl side of basic residues at the NH2-terminus of peptides. 2D electrophoresis (2DE) was used to show that Ap-B is modified by phosphorylation. Detection of Ap-B by western blot after 2DE reveals several isoforms with different isoelectric points. Using alkaline phosphatase, Pro-Q Diamond phosphorylation-specific dye and kinase-specific inhibitors, we confirmed that Ap-B is phosphorylated. Phosphorylation can alter the structure of proteins leading to changes in their activity, localization, stability and association with other interacting molecules. We showed that Ap-B phosphorylation might delay its turnover. Our study illustrates the central role of the crosstalk between kinases and proteases in the regulation of many biological processes.
Collapse
|
2
|
Liu Y, Bockermann R, Hadi M, Safari I, Carrion B, Kveiborg M, Issazadeh-Navikas S. ADAM12 is a costimulatory molecule that determines Th1 cell fate and mediates tissue inflammation. Cell Mol Immunol 2021; 18:1904-1919. [PMID: 32572163 PMCID: PMC8322154 DOI: 10.1038/s41423-020-0486-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/31/2020] [Indexed: 12/18/2022] Open
Abstract
A disintegrin and metalloproteinase (ADAM)12 was previously found to be expressed in T cells in the inflamed brain. However, the function of ADAM12 in T-cell responses in general and in tissue inflammation has not been examined. Here, we studied the role of ADAM12 in T-cell responses, fate determination on activation, and its functions in T cells to mediate tissue inflammation. We identified ADAM12 as a costimulatory molecule that is expressed on naive T cells and downregulated on stimulation. ADAM12 mimics CD28 costimulatory signaling to activate and induce the proliferation of T-helper 1 (Th1) cells. Monoclonal ADAM12 Fab antibodies trigger T-cell activation by amplifying TCR signaling to stimulate T-bet-mediated IFNγ production. Lack of genomic ADAM12 and its knockdown in T cells diminished T-bet and IFNγ production in Th1 cells, whereas other T cells, including Th17 cells, were unaffected. ADAM12 had similar functions in vivo on myelin antigen (MOG35-55)-induced T-cell activation. We found that genetic loss of ADAM12 profoundly alleviated Th1-mediated neuroinflammation and thus disease severity in experimental autoimmune encephalomyelitis, a model of multiple sclerosis. Transcriptomic profiling of MOG35-55-specific ADAM12-/- T cells revealed differentially expressed genes that are important for T-cell activation, proliferation, and costimulatory signaling and Th1 pathogenicity, consistent with their inability to cause T-cell-mediated skin inflammation in a model of adoptive delayed-type hypersensitivity. We conclude that ADAM12 is a T-cell costimulatory molecule that contributes to the pathogenesis of tissue inflammation and a potential target for the treatment of Th1-mediated diseases.
Collapse
Affiliation(s)
- Yawei Liu
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Robert Bockermann
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Mahdieh Hadi
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Iman Safari
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Belinda Carrion
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Marie Kveiborg
- BRIC, Faculty of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Shohreh Issazadeh-Navikas
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark.
| |
Collapse
|
3
|
Ogawa K, Lin Q, Li L, Bai X, Chen X, Chen H, Kong R, Wang Y, Zhu H, He F, Xu Q, Liu L, Li M, Zhang S, Nagaoka K, Carlson R, Safran H, Charpentier K, Sun B, Wands J, Dong X. Aspartate β-hydroxylase promotes pancreatic ductal adenocarcinoma metastasis through activation of SRC signaling pathway. J Hematol Oncol 2019; 12:144. [PMID: 31888763 PMCID: PMC6937817 DOI: 10.1186/s13045-019-0837-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 12/11/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Signaling pathways critical for embryonic development re-emerge in adult pancreas during tumorigenesis. Aspartate β-hydroxylase (ASPH) drives embryonic cell motility/invasion in pancreatic development/differentiation. We explored if dysregulated ASPH is critically involved in pancreatic cancer pathogenesis. METHODS To demonstrate if/how ASPH mediates malignant phenotypes, proliferation, migration, 2-D/3-D invasion, pancreatosphere formation, immunofluorescence, Western blot, co-immunoprecipitation, invadopodia formation/maturation/function, qRT-PCR, immunohistochemistry (IHC), and self-developed in vitro metastasis assays were performed. Patient-derived xenograft (PDX) models of human pancreatic ductal adenocarcinoma (PDAC) were established to illustrate in vivo antitumor effects of the third-generation small molecule inhibitor specifically against ASPH's β-hydroxylase activity. Prognostic values of ASPH network components were evaluated with Kaplan-Meier plots, log-rank tests, and Cox proportional hazards regression models. RESULTS ASPH renders pancreatic cancer cells more aggressive phenotypes characterized by epithelial-mesenchymal transition (EMT), 2-D/3-D invasion, invadopodia formation/function as demonstrated by extracellular matrix (ECM) degradation, stemness (cancer stem cell marker upregulation and pancreatosphere formation), transendothelial migration (mimicking intravasation/extravasation), and sphere formation (mimicking metastatic colonization/outgrowth at distant sites). Mechanistically, ASPH activates SRC cascade through direct physical interaction with ADAM12/ADAM15 independent of FAK. The ASPH-SRC axis enables invadopodia construction and initiates MMP-mediated ECM degradation/remodeling as executors for invasiveness. Pharmacologic inhibition of invadopodia attenuates in vitro metastasis. ASPH fosters primary tumor development and pulmonary metastasis in PDX models of PDAC, which is blocked by a leading compound specifically against ASPH enzymatic activity. ASPH is silenced in normal pancreas, progressively upregulated from pre-malignant lesions to invasive/advanced stages of PDAC. Expression profiling of ASPH-SRC network components independently/jointly predicts clinical outcome of PDAC patients. Compared to a negative-low level, a moderate-very high level of ASPH, ADAM12, activated SRC, and MMPs correlated with curtailed overall survival (OS) of pancreatic cancer patients (log-rank test, ps < 0.001). The more unfavorable molecules patients carry, the more deleterious prognosis is destinated. Patients with 0-2 (n = 4), 3-5 (n = 8), 6-8 (n = 24), and 9-12 (n = 73) unfavorable expression scores of the 5 molecules had median survival time of 55.4, 15.9, 9.7, and 5.0 months, respectively (p < 0.001). CONCLUSION Targeting the ASPH-SRC axis, which is essential for propagating multi-step PDAC metastasis, may specifically/substantially retard development/progression and thus improve prognosis of PDAC.
Collapse
Affiliation(s)
- Kosuke Ogawa
- Liver Research Center, Rhode Island Hospital, Warren Alpert Medical School, Brown University, 55 Claverick Street, 4th Fl., Providence, RI, 02903, USA
| | - Qiushi Lin
- Department of Internal Medicine, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731014, USA
| | - Le Li
- Department of Pancreatic and Biliary Surgery; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China
| | - Xuewei Bai
- Liver Research Center, Rhode Island Hospital, Warren Alpert Medical School, Brown University, 55 Claverick Street, 4th Fl., Providence, RI, 02903, USA.,Department of Pancreatic and Biliary Surgery; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China
| | - Xuesong Chen
- Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150040, Heilongjiang Province, People's Republic of China
| | - Hua Chen
- Department of Pancreatic and Biliary Surgery; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China
| | - Rui Kong
- Department of Pancreatic and Biliary Surgery; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China
| | - Yongwei Wang
- Department of Pancreatic and Biliary Surgery; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China
| | - Hong Zhu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, People's Republic of China
| | - Fuliang He
- Department of Internal Medicine, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731014, USA.,Department of Interventional Therapy, Beijing Shijitan Hospital, Capital Medical University, The 9th Affiliated Hospital of Peking University, Beijing, People's Republic of China
| | - Qinggang Xu
- Department of Internal Medicine, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731014, USA.,Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, People's Republic of China
| | - Lianxin Liu
- Department of Hepatic Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, The University of Sciences and Technology of China, No. 17 Lujiang Road, Hefei City, 230001, An Hui Province, People's Republic of China
| | - Min Li
- Immunobiology & Transplant Science Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Songhua Zhang
- Liver Research Center, Rhode Island Hospital, Warren Alpert Medical School, Brown University, 55 Claverick Street, 4th Fl., Providence, RI, 02903, USA
| | - Katsuya Nagaoka
- Liver Research Center, Rhode Island Hospital, Warren Alpert Medical School, Brown University, 55 Claverick Street, 4th Fl., Providence, RI, 02903, USA
| | - Rolf Carlson
- Liver Research Center, Rhode Island Hospital, Warren Alpert Medical School, Brown University, 55 Claverick Street, 4th Fl., Providence, RI, 02903, USA
| | - Howard Safran
- Division of Hematology/Oncology, Rhode Island Hospital/The Miriam Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Kevin Charpentier
- Department of Surgery, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang Province, People's Republic of China
| | - Jack Wands
- Liver Research Center, Rhode Island Hospital, Warren Alpert Medical School, Brown University, 55 Claverick Street, 4th Fl., Providence, RI, 02903, USA.
| | - Xiaoqun Dong
- Liver Research Center, Rhode Island Hospital, Warren Alpert Medical School, Brown University, 55 Claverick Street, 4th Fl., Providence, RI, 02903, USA. .,Department of Internal Medicine, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 731014, USA. .,Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
4
|
Seo T, Sakon T, Nakazawa S, Nishioka A, Watanabe K, Matsumoto K, Akasaka M, Shioi N, Sawada H, Araki S. Haemorrhagic snake venom metalloproteases and human
ADAM
s cleave
LRP
5/6, which disrupts cell–cell adhesions
in vitro
and induces haemorrhage
in vivo. FEBS J 2017; 284:1657-1671. [DOI: 10.1111/febs.14066] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/11/2017] [Accepted: 03/22/2017] [Indexed: 11/25/2022]
Affiliation(s)
- Tadahiko Seo
- Sugashima Marine Biological Laboratory Graduate School of Science Nagoya University Japan
| | - Taketo Sakon
- Sugashima Marine Biological Laboratory Graduate School of Science Nagoya University Japan
| | - Shiori Nakazawa
- Sugashima Marine Biological Laboratory Graduate School of Science Nagoya University Japan
| | - Asuka Nishioka
- Sugashima Marine Biological Laboratory Graduate School of Science Nagoya University Japan
| | - Kohei Watanabe
- Sugashima Marine Biological Laboratory Graduate School of Science Nagoya University Japan
| | - Kaori Matsumoto
- Sugashima Marine Biological Laboratory Graduate School of Science Nagoya University Japan
| | - Mari Akasaka
- Sugashima Marine Biological Laboratory Graduate School of Science Nagoya University Japan
| | - Narumi Shioi
- Department of Chemistry Faculty of Science Fukuoka University Japan
| | - Hitoshi Sawada
- Sugashima Marine Biological Laboratory Graduate School of Science Nagoya University Japan
| | - Satohiko Araki
- Sugashima Marine Biological Laboratory Graduate School of Science Nagoya University Japan
| |
Collapse
|
5
|
Chen YJ, Liu WH, Chang LS. Hydroquinone-induced FOXP3-ADAM17-Lyn-Akt-p21 signaling axis promotes malignant progression of human leukemia U937 cells. Arch Toxicol 2017; 91:983-997. [PMID: 27307158 DOI: 10.1007/s00204-016-1753-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/08/2016] [Indexed: 12/17/2022]
Abstract
Hydroquinone (1,4-benzenediol; HQ), a major marrow metabolite of the leukemogen benzene, has been proven to evoke benzene-related hematological disorders and myelotoxicity in vitro and in vivo. The goal of the present study was to explore the role of FOXP3 in HQ-induced malignant progression of U937 human leukemia cells. U937 cells were treated with 5 μM HQ for 24 h, and the cells were re-suspended in serum-containing medium without HQ for 2 days. The same procedure was repeated three times, and the resulting U937/HQ cells were maintained in cultured medium containing 5 μM HQ. Proliferation and colony formation of U937/HQ cells were notably higher than those of U937 cells. Ten-eleven translocation methylcytosine dioxygenase-mediated demethylation of the Treg-specific demethylated region in FOXP3 gene resulted in higher FOXP3 expression in U937/HQ cells than in U937 cells. FOXP3-induced miR-183 expression reduced β-TrCP mRNA stability and suppressed β-TrCP-mediated Sp1 degradation, leading to up-regulation of Sp1 expression in U937/HQ cells. Sp1 up-regulation further increased ADAM17 and Lyn expression, and ADAM17 up-regulation stimulated Lyn activation in U937/HQ cells. Moreover, U937/HQ cells showed higher Lyn-mediated Akt activation and cytoplasmic p21 expression than U937 cells did. Abolishment of Akt activation decreased cytoplasmic p21 expression in U937/HQ cells. Suppression of FOXP3, ADAM17, and Lyn expression, as well as Akt inactivation, repressed proliferation and clonogenicity of U937/HQ cells. Together with the finding that cytoplasmic p21 shows anti-apoptotic and oncogenic activities in cancer cells, the present data suggest a role of FOXP3/ADAM17/Lyn/Akt/p21 signaling axis in HQ-induced hematological disorders.
Collapse
Affiliation(s)
- Ying-Jung Chen
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Wen-Hsin Liu
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Long-Sen Chang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan.
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| |
Collapse
|
6
|
Cipriani P, Di Benedetto P, Ruscitti P, Liakouli V, Berardicurti O, Carubbi F, Ciccia F, Guggino G, Zazzeroni F, Alesse E, Triolo G, Giacomelli R. Perivascular Cells in Diffuse Cutaneous Systemic Sclerosis Overexpress Activated ADAM12 and Are Involved in Myofibroblast Transdifferentiation and Development of Fibrosis. J Rheumatol 2016; 43:1340-9. [DOI: 10.3899/jrheum.150996] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2016] [Indexed: 02/06/2023]
Abstract
Objective.Microvascular damage is pivotal in the pathogenesis of systemic sclerosis (SSc), preceding fibrosis, and whose trigger is not still fully understood. Perivascular progenitor cells, with profibrotic activity and function, are identified by the expression of the isoform 12 of ADAM (ADAM12) and this molecule may be upregulated by transforming growth factor-β (TGF-β). The goal of this work was to evaluate whether pericytes in the skin of patients with diffuse cutaneous SSc (dcSSc) expressed ADAM12, suggesting their potential contribution to the fibrotic process, and whether TGF-β might modulate this molecule.Methods.After ethical approval, mesenchymal stem cells (MSC) and fibroblasts (FB) were isolated from bone marrow and skin samples collected from 20 patients with dcSSc. ADAM12 expression was investigated in the skin and in isolated MSC and FB treated with TGF-β by immunofluorescence, quantitative real-time PCR, and western blot. Further, we silenced ADAM12 expression in both dcSSc-MSC and -FB to confirm the TGF-β modulation.Results.Pericytes and FB of dcSSc skin showed an increased expression of ADAM12 when compared with healthy control skin. TGF-β in vitro treatment induced a significant increase of ADAM12 in both SSc-MSC and -FB, with the higher levels observed in dcSSc cells. After ADAM12 silencing, the TGF-β ability to upregulate α-smooth muscle actin in both SSc-MSC and SSc-FB was inhibited.Conclusion.Our results suggest that in SSc, pericytes that transdifferentiate toward activated FB are present in the vascular tree, and TGF-β, while increasing ADAM12 expression, may modulate this transdifferentiation.
Collapse
|
7
|
The Disintegrin and Metalloprotease ADAM12 Is Associated with TGF-β-Induced Epithelial to Mesenchymal Transition. PLoS One 2015; 10:e0139179. [PMID: 26407179 PMCID: PMC4583281 DOI: 10.1371/journal.pone.0139179] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 09/08/2015] [Indexed: 12/21/2022] Open
Abstract
The increased expression of the Disintegrin and Metalloprotease ADAM12 has been associated with human cancers, however its role remain unclear. We have previously reported that ADAM12 expression is induced by the transforming growth factor, TGF-β and promotes TGF-β-dependent signaling through interaction with the type II receptor of TGF-β. Here we explore the implication of ADAM12 in TGF-β-mediated epithelial to mesenchymal transition (EMT), a key process in cancer progression. We show that ADAM12 expression is correlated with EMT markers in human breast cancer cell lines and biopsies. Using a non-malignant breast epithelial cell line (MCF10A), we demonstrate that TGF-β-induced EMT increases expression of the membrane-anchored ADAM12L long form. Importantly, ADAM12L overexpression in MCF10A is sufficient to induce loss of cell-cell contact, reorganization of actin cytoskeleton, up-regulation of EMT markers and chemoresistance. These effects are independent of the proteolytic activity but require the cytoplasmic tail and are specific of ADAM12L since overexpression of ADAM12S failed to induce similar changes. We further demonstrate that ADAM12L-dependent EMT is associated with increased phosphorylation of Smad3, Akt and ERK proteins. Conversely, inhibition of TGF-β receptors or ERK activities reverses ADAM12L-induced mesenchymal phenotype. Together our data demonstrate that ADAM12L is associated with EMT and contributes to TGF-β-dependent EMT by favoring both Smad-dependent and Smad-independent pathways.
Collapse
|
8
|
Aghababaei M, Beristain AG. The Elsevier Trophoblast Research Award Lecture: Importance of metzincin proteases in trophoblast biology and placental development: a focus on ADAM12. Placenta 2015; 36 Suppl 1:S11-9. [PMID: 25589360 DOI: 10.1016/j.placenta.2014.12.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/04/2014] [Accepted: 12/17/2014] [Indexed: 10/25/2022]
Abstract
Placental development is a highly regulated process requiring signals from both fetal and maternal uterine compartments. Within this complex system, trophoblasts, placental cells of epithelial lineage, form the maternal-fetal interface controlling nutrient, gas and waste exchange. The commitment of progenitor villous cytotrophoblasts to differentiate into diverse trophoblast subsets is a fundamental process in placental development. Differentiation of trophoblasts into invasive stromal- and vascular-remodeling subtypes is essential for uterine arterial remodeling and placental function. Inadequate placentation, characterized by defects in trophoblast differentiation, may underlie the earliest cellular events driving pregnancy disorders such as preeclampsia and fetal growth restriction. Molecularly, invasive trophoblasts acquire characteristics defined by profound alterations in cell-cell and cell-matrix adhesion, cytoskeletal reorganization and production of proteolytic factors. To date, most studies have investigated the importance of the matrix metalloproteinases (MMPs) and their ability to efficiently remodel components of the extracellular matrix (ECM). However, it is now becoming clear that besides MMPs, other related proteases regulate trophoblast invasion via mechanisms other than ECM turnover. In this review, we will summarize the current knowledge on the regulation of trophoblast invasion by members of the metzincin family of metalloproteinases. Specifically, we will discuss the emerging roles that A Disintegrin and Metalloproteinases (ADAMs) play in placental development, with a particular focus on the ADAM subtype, ADAM12.
Collapse
Affiliation(s)
- Mahroo Aghababaei
- Department of Obstetrics and Gynecology, The University of British Columbia, Canada; The Child and Family Research Institute, Vancouver, Canada
| | - Alexander G Beristain
- Department of Obstetrics and Gynecology, The University of British Columbia, Canada; The Child and Family Research Institute, Vancouver, Canada.
| |
Collapse
|
9
|
Ebsen H, Lettau M, Kabelitz D, Janssen O. Subcellular localization and activation of ADAM proteases in the context of FasL shedding in T lymphocytes. Mol Immunol 2015; 65:416-28. [PMID: 25745808 DOI: 10.1016/j.molimm.2015.02.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 01/20/2015] [Accepted: 02/08/2015] [Indexed: 10/23/2022]
Abstract
The "A Disintegrin And Metalloproteinases" (ADAMs) form a subgroup of the metzincin endopeptidases. Proteolytically active members of this protein family act as sheddases and govern key processes in development and inflammation by regulating cell surface expression and release of cytokines, growth factors, adhesion molecules and their receptors. In T lymphocytes, ADAM10 sheds the death factor Fas Ligand (FasL) and thereby regulates T cell activation, death and effector function. Although FasL shedding by ADAM10 was confirmed in several studies, its regulation is still poorly defined. We recently reported that ADAM10 is highly abundant on T cells whereas its close relative ADAM17 is expressed at low levels and transiently appears at the cell surface upon stimulation. Since FasL is also stored intracellularly and brought to the plasma membrane upon stimulation, we addressed where the death factor gets exposed to ADAM proteases. We report for the first time that both ADAM10 and ADAM17 are associated with FasL-containing secretory lysosomes. Moreover, we demonstrate that TCR/CD3/CD28-stimulation induces a partial positioning of both proteases and FasL to lipid rafts and only the activation-induced raft-positioning results in FasL processing. TCR/CD3/CD28-induced FasL proteolysis is markedly affected by reducing both ADAM10 and ADAM17 protein levels, indicating that in human T cells also ADAM17 is implicated in FasL processing. Since FasL shedding is affected by cholesterol depletion and by inhibition of Src kinases or palmitoylation, we conclude that it requires mobilization and co-positioning of ADAM proteases in lipid raft-like platforms associated with an activation of raft-associated Src-family kinases.
Collapse
Affiliation(s)
- Henriette Ebsen
- University of Kiel, Institute of Immunology, University Hospital Schleswig-Holstein Campus Kiel, Arnold-Heller-Str. 3 Bldg 17, D-24105 Kiel, Germany
| | - Marcus Lettau
- University of Kiel, Institute of Immunology, University Hospital Schleswig-Holstein Campus Kiel, Arnold-Heller-Str. 3 Bldg 17, D-24105 Kiel, Germany
| | - Dieter Kabelitz
- University of Kiel, Institute of Immunology, University Hospital Schleswig-Holstein Campus Kiel, Arnold-Heller-Str. 3 Bldg 17, D-24105 Kiel, Germany
| | - Ottmar Janssen
- University of Kiel, Institute of Immunology, University Hospital Schleswig-Holstein Campus Kiel, Arnold-Heller-Str. 3 Bldg 17, D-24105 Kiel, Germany.
| |
Collapse
|
10
|
Ebsen H, Lettau M, Kabelitz D, Janssen O. Identification of SH3 domain proteins interacting with the cytoplasmic tail of the a disintegrin and metalloprotease 10 (ADAM10). PLoS One 2014; 9:e102899. [PMID: 25036101 PMCID: PMC4103893 DOI: 10.1371/journal.pone.0102899] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 06/25/2014] [Indexed: 11/19/2022] Open
Abstract
The a disintegrin and metalloproteases (ADAMs) play a pivotal role in the control of development, adhesion, migration, inflammation and cancer. Although numerous substrates of ADAM10 have been identified, the regulation of its surface expression and proteolytic activity is still poorly defined. One current hypothesis is that both processes are in part modulated by protein-protein interactions mediated by the intracellular portion of the protease. For related proteases, especially proline-rich regions serving as docking sites for Src homology domain 3 (SH3) domain-containing proteins proved to be important for mediating regulatory interactions. In order to identify ADAM10-binding SH3 domain proteins, we screened the All SH3 Domain Phager library comprising 305 human SH3 domains using a GST fusion protein with the intracellular region of human ADAM10 as a bait for selection. Of a total of 291 analyzed phage clones, we found 38 SH3 domains that were precipitated with the ADAM10-derived fusion protein but not with GST. We verified the binding to the cytosolic portion of ADAM10 for several candidates by co-immunoprecipitation and/or pull down analyses. Intriguingly, several of the identified proteins have been implicated in regulating surface appearance and/or proteolytic activity of related ADAMs. Thus, it seems likely that they also play a role in ADAM10 biology.
Collapse
Affiliation(s)
- Henriette Ebsen
- University of Kiel, Molecular Immunology, Institute for Immunology, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Marcus Lettau
- University of Kiel, Molecular Immunology, Institute for Immunology, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Dieter Kabelitz
- University of Kiel, Molecular Immunology, Institute for Immunology, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Ottmar Janssen
- University of Kiel, Molecular Immunology, Institute for Immunology, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
- * E-mail:
| |
Collapse
|
11
|
Rao VH, Vogel K, Yanagida JK, Marwaha N, Kandel A, Trempus C, Repertinger SK, Hansen LA. Erbb2 up-regulation of ADAM12 expression accelerates skin cancer progression. Mol Carcinog 2014; 54:1026-36. [PMID: 24798404 DOI: 10.1002/mc.22171] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/31/2014] [Accepted: 04/07/2014] [Indexed: 12/17/2022]
Abstract
Solar ultraviolet (UV) radiation can cause severe damage to the skin and is the primary cause of most skin cancer. UV radiation causes DNA damage leading to mutations and also activates the Erbb2/HER2 receptor through indirect mechanisms involving reactive oxygen species. We hypothesized that Erbb2 activation accelerates the malignant progression of UV-induced skin cancer. Following the induction of benign squamous papillomas by UV exposure of v-ras(Ha) transgenic Tg.AC mice, mice were treated topically with the Erbb2 inhibitor AG825 and tumor progression monitored. AG825 treatment reduced tumor volume, increased tumor regression, and delayed the development of malignant squamous cell carcinoma (SCC). Progression to malignancy was associated with increased Erbb2 and ADAM12 (A Disintegin And Metalloproteinase 12) transcripts and protein, while inhibition of Erbb2 blocked the increase in ADAM12 message upon malignant progression. Similarly, human SCC and SCC cell lines had increased ADAM12 protein and transcripts when compared to normal controls. To determine whether Erbb2 up-regulation of ADAM12 contributed to malignant progression of skin cancer, Erbb2 expression was modulated in cultured SCC cells using forced over-expression or siRNA targeting, demonstrating up-regulation of ADAM12 by Erbb2. Furthermore, ADAM12 transfection or siRNA targeting revealed that ADAM12 increased both the migration and invasion of cutaneous SCC cells. Collectively, these results suggest Erbb2 up-regulation of ADAM12 as a novel mechanism contributing to the malignant progression of UV-induced skin cancer. Inhibition of Erbb2/HER2 reduced tumor burden, increased tumor regression, and delayed the progression of benign skin tumors to malignant SCC in UV-exposed mice. Inhibition of Erbb2 suppressed the increase in metalloproteinase ADAM12 expression in skin tumors, which in turn increased migration and tumor cell invasiveness.
Collapse
Affiliation(s)
- Velidi H Rao
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska
| | - Kristen Vogel
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska
| | - Jodi K Yanagida
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska
| | - Nitin Marwaha
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska
| | - Amrit Kandel
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska
| | - Carol Trempus
- Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | | | - Laura A Hansen
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska
| |
Collapse
|
12
|
Nyren-Erickson EK, Bouton M, Raval M, Totzauer J, Mallik S, Alberto N. Urinary concentrations of ADAM 12 from breast cancer patients pre- and post-surgery vs. cancer-free controls: a clinical study for biomarker validation. J Negat Results Biomed 2014; 13:5. [PMID: 24690292 PMCID: PMC3977887 DOI: 10.1186/1477-5751-13-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 03/18/2014] [Indexed: 02/07/2023] Open
Abstract
Background The ADAMs (A Disintegrin and Metalloproteinases) are a family of multi-domain, zinc-dependent metalloproteinase enzymes. ADAM 12 has been previously associated with the onset and progression of breast cancer, and elevated levels of ADAM 12 have been previously found in the urine of breast cancer patients. Aims of the current study are: 1) establish the viability of urinary ADAM 12 as a diagnostic marker for breast cancer, and 2) explore the effects of surgical tumor removal on the levels of urinary ADAM 12. Methods A total of 96 patients have been recruited for this study, including 50 patients diagnosed with cancer, and 46 age-matched controls. Commercially available ELISA kits for ADAM 12 were used to quantify the presence and concentration of this enzyme in the urine from cancer patients with ductal carcinoma in situ (DCIS) and invasive breast cancer (IBC) both prior to any treatment and approximately two weeks following surgery, as well as from controls. Results We find no statistically significant differences between the concentrations of ADAM 12 in the urine of breast cancer patients prior to treatment and that of their age-matched controls; however the concentration of ADAM 12, both alone and as a function of urine total protein, are significantly elevated following surgery (p < 0.0001). Patients who underwent a mastectomy have significantly higher urinary ADAM 12 concentrations than those who underwent a lumpectomy (significant at p = 0.0271). Conclusions These findings suggest that urinary ADAM 12 may not correlate directly with the status and stage of breast cancer as previously thought; rather these increases may be a result of tissue injury and inflammation from biopsy and surgical resection. Results of this study may suggest a need for biomarkers to be evaluated carefully in the context of tissue damage.
Collapse
Affiliation(s)
| | | | | | | | | | - Neville Alberto
- Roger Maris Cancer Center, Sanford Health, Fargo, ND 58122, USA.
| |
Collapse
|
13
|
Coles CA, Wadeson J, Knight MI, Cafe LM, Johns WH, White JD, Greenwood PL, McDonagh MB. A disintegrin and metalloprotease-12 is type I myofiber specific in Bos taurus and Bos indicus cattle. J Anim Sci 2014; 92:1473-83. [PMID: 24663211 DOI: 10.2527/jas.2013-7069] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A disintegrin and metalloproteinase-12 (ADAM12) is involved in the regulation of myogenesis and adipogenesis and is of interest as a potential target to manipulate skeletal muscle development and intramuscular fat (IMF) deposition in cattle to increase beef yield and improve meat quality. The longissimus thoracis muscle (LM) and semitendinosus muscle (STM) from 5 Bos taurus (Angus) and 5 Bos indicus (Brahman) cattle were collected for histological and ADAM12 gene and protein expression analysis. Myofiber typing was used to determine if ADAM12 expression patterns related to differences in muscling and IMF deposition, which are influenced by proportions of the different myofiber types. The STM was found to contain a higher proportion of glycolytic myofibers than the LM, which contained a greater proportion of oxidative myofibers (myofiber ratio of glycolytic to more oxidative types in LM and STM of 1.1 and 3.5, respectively; P < 0.05). ADAM12 gene expression, fluorescent immunohistochemical staining for ADAM12, and image analysis found ADAM12 to be greater in the LM (P < 0.05). Regression analysis found a strong, positive relationship for the distribution of ADAM12 against the proportion of type I myofibers (P < 0.05, r(2) = 0.86). These findings suggest ADAM12 is upregulated in muscles with more slow-oxidative myofibres, such as the LM, and is linked to type I myofibers in cattle. ADAM12 may be important in the regulation and maintenance slow myofibers in the LM muscle.
Collapse
Affiliation(s)
- C A Coles
- Victorian Department of Environment and Primary Industries, Bundoora, VIC 3083, Australia
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Chen J, Jiang X, Duan Y, Long J, Bartsch JW, Deng L. ADAM8 in asthma. Friend or foe to airway inflammation? Am J Respir Cell Mol Biol 2014; 49:875-84. [PMID: 23837412 DOI: 10.1165/rcmb.2013-0168tr] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Airway inflammation has been suggested as the pathological basis in asthma pathogenesis. Recruitment of leukocytes from the vasculature into airway sites is essential for induction of airway inflammation, a process thought to be mediated by a disintegrin and metalloprotease 8 (ADAM8). However, there is an apparent controversy about whether ADAM8 helps or hampers transmigration of leukocytes through endothelium in airway inflammation of asthma. This review outlines the current contradictory concepts concerning the role of ADAM8 in airway inflammation, particularly focusing on the recruitment of leukocytes during asthma, and attempts to bridge the existing experimental data on the basis of the functional analysis of different domains of ADAM8 and their endogenous processing in vivo. We suggest a possible hypothesis for the specific mechanism by which ADAM8 regulates the transmigration of leukocytes to explain the disparity existing in current studies, and we also raise some questions that require future investigations.
Collapse
Affiliation(s)
- Jun Chen
- 1 Key Lab of Biorheological Science and Technology, Ministry of Education, "National 985 Project" Institute of Biorheology and Gene Regulation, Bioengineering College, Chongqing University, Chongqing, P.R. China
| | | | | | | | | | | |
Collapse
|
15
|
Aghababaei M, Perdu S, Irvine K, Beristain AG. A disintegrin and metalloproteinase 12 (ADAM12) localizes to invasive trophoblast, promotes cell invasion and directs column outgrowth in early placental development. Mol Hum Reprod 2013; 20:235-49. [PMID: 24243624 DOI: 10.1093/molehr/gat084] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
During pregnancy, stromal- and vascular-remodeling trophoblasts serve critical roles in directing placental development acquiring pro-invasive characteristics. The A Disintegrin and Metalloproteinase (ADAM) family of multifunctional proteins direct cellular processes across multiple organ systems via their intrinsic catalytic, cell adhesive and intracellular signaling properties. ADAM12, existing as two distinct splice variants (ADAM12L and ADAM12S), is highly expressed in the human placenta and promotes cell migration and invasion in several tumor cell lines; however, its role in trophoblast biology is unknown. In this study, ADAM12 was localized to anchoring trophoblast columns in first trimester placentas and to highly invasive extracellular matrix-degrading trophoblasts in placental villous explants. The importance of ADAM12 in directing trophoblast invasion was tested using loss-of and gain-of-function strategies, where siRNA-directed knockdown of ADAM12 inhibited trophoblast cell invasion while over-expression promoted migration and invasion in two trophoblastic cell models. In placental villous explant cultures, siRNA-directed loss of ADAM12 significantly dampened trophoblast column outgrowth. Additionally, we provide functional evidence for the ADAM12S variant in promoting trophoblast invasion and column outgrowth through a mechanism requiring its catalytic activity. This is the first study to assign a function for ADAM12 in trophoblast biology, where ADAM12 may play a central role regulating the behavior of invasive trophoblast subsets in early pregnancy. This study also underlines the importance of ADAM12L and ADAM12S in directing cell motility in normal developmental processes outside of cancer, specifically highlighting a potentially important function of ADAM12S in directing early placental development.
Collapse
Affiliation(s)
- M Aghababaei
- Department of Obstetrics and Gynecology, The University of British Columbia, Vancouver, Canada
| | | | | | | |
Collapse
|
16
|
ADAM12 is expressed in the tumour vasculature and mediates ectodomain shedding of several membrane-anchored endothelial proteins. Biochem J 2013; 452:97-109. [PMID: 23458101 DOI: 10.1042/bj20121558] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
ADAM (a disintegrin and metalloproteinase) 12 is a metalloprotease implicated in cancer progression. ADAM12 can activate membrane-anchored proteins, such as sonic hedgehog, Delta-like 1 and certain epidermal growth factor receptor ligands, through a process called ectodomain shedding. We screened several membrane-anchored proteins to further dissect the substrate profile of ADAM12-mediated ectodomain shedding, and found shedding of five previously unreported substrates [Kitl1, VE-cadherin (vascular endothelial cadherin), Flk-1 (fetal liver kinase 1), Tie-2, and VCAM-1 (vascular cell adhesion molecule 1)], of which the latter four are specifically expressed by endothelial cells. We also observed that ADAM12 expression was increased in the tumour vasculature of infiltrating ductal carcinoma of the human breast as compared with little to no expression in normal breast tissue vasculature, suggesting a role for ADAM12 in tumour vessels. These results prompted us to further evaluate ADAM12-mediated shedding of two endothelial cell proteins, VE-cadherin and Tie-2. Endogenous ADAM12 expression was very low in cultured endothelial cells, but was significantly increased by cytokine stimulation. In parallel, the shed form of VE-cadherin was elevated in such cytokine-stimulated endothelial cells, and ADAM12 siRNA (small interfering RNA) knockdown reduced cytokine-induced shedding of VE-cadherin. In conclusion, the results of the present study demonstrate a role for ADAM12 in ectodomain shedding of several membrane-anchored endothelial proteins. We speculate that this process may have importance in tumour neovascularization or/and tumour cell extravasation.
Collapse
|
17
|
Díaz B, Yuen A, Iizuka S, Higashiyama S, Courtneidge SA. Notch increases the shedding of HB-EGF by ADAM12 to potentiate invadopodia formation in hypoxia. ACTA ACUST UNITED AC 2013; 201:279-92. [PMID: 23589494 PMCID: PMC3628517 DOI: 10.1083/jcb.201209151] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hypoxia increases the levels of ADAM12 in a Notch-dependent manner, leading to increased ectodomain shedding of HB-EGF and subsequent promotion of invadopodia formation. Notch regulates cell–cell contact-dependent signaling and is activated by hypoxia, a microenvironmental condition that promotes cellular invasion during both normal physiology and disease. The mechanisms by which hypoxia and Notch regulate cellular invasion are not fully elucidated. In this paper, we show that, in cancer cells, hypoxia increased the levels and activity of the ADAM12 metalloprotease in a Notch signaling–dependent manner, leading to increased ectodomain shedding of the epidermal growth factor (EGF) receptor (EGFR) ligand heparin-binding EGF-like growth factor. Released HB-EGF induced the formation of invadopodia, cellular structures that aid cancer cell invasion. Thus, we describe a signaling pathway that couples cell contact–dependent signaling with the paracrine activation of the EGFR, indicating cross talk between the Notch and EGFR pathways in promoting cancer cell invasion. This signaling pathway might regulate the coordinated acquisition of invasiveness by neighboring cells and mediate the communication between normoxic and hypoxic areas of tumors to facilitate cancer cell invasion.
Collapse
Affiliation(s)
- Begoña Díaz
- Cancer Center, Tumor Microenvironment Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA.
| | | | | | | | | |
Collapse
|
18
|
Nyren-Erickson EK, Jones JM, Srivastava DK, Mallik S. A disintegrin and metalloproteinase-12 (ADAM12): function, roles in disease progression, and clinical implications. Biochim Biophys Acta Gen Subj 2013; 1830:4445-55. [PMID: 23680494 DOI: 10.1016/j.bbagen.2013.05.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 05/03/2013] [Accepted: 05/06/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND A disintegrin and metalloproteinase-12 (ADAM12) is a member of the greater ADAM family of enzymes: these are multifunctional, generally membrane-bound, zinc proteases for which there are forty genes known (21 of these appearing in humans). ADAM12 has been implicated in the pathogenesis of various cancers, liver fibrogenesis, hypertension, and asthma, and its elevation or decrease in human serum has been linked to these and other physiological/pathological conditions. SCOPE In this review, we begin with a brief overview of the ADAM family of enzymes and protein structure. We then discuss the role of ADAM12 in the progression and/or diagnosis of various disease conditions, and we will conclude with an exploration of currently known natural and synthetic inhibitors. MAJOR CONCLUSION ADAM12 has potential to emerge as a successful drug target, although targeting the metalloproteinase domain with any specificity will be difficult to achieve due to structural similarity between the members of the ADAM and MMP family of enzymes. Overall, more research is required to establish ADAM12 being as a highly desirable biomarker and drug target of different diseases, and their selective inhibitors as potential therapeutic agents. GENERAL SIGNIFICANCE Given the appearance of elevated levels of ADAM12 in various diseases, particularly breast cancer, our understanding of this enzyme both as a biomarker and a potential drug target could help make significant inroads into both early diagnosis and treatment of disease.
Collapse
Affiliation(s)
- Erin K Nyren-Erickson
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58108-6050, USA
| | | | | | | |
Collapse
|
19
|
Kim JH, Lee YG, Yoo S, Oh J, Jeong D, Song WK, Yoo BC, Rhee MH, Park J, Cha SH, Hong S, Cho JY. Involvement of Src and the actin cytoskeleton in the antitumorigenic action of adenosine dialdehyde. Biochem Pharmacol 2013; 85:1042-56. [DOI: 10.1016/j.bcp.2013.01.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 12/28/2012] [Accepted: 01/18/2013] [Indexed: 01/06/2023]
|
20
|
Stautz D, Leyme A, Grandal MV, Albrechtsen R, van Deurs B, Wewer U, Kveiborg M. Cell-surface metalloprotease ADAM12 is internalized by a clathrin- and Grb2-dependent mechanism. Traffic 2012; 13:1532-46. [PMID: 22882974 DOI: 10.1111/j.1600-0854.2012.01405.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 08/08/2012] [Accepted: 08/10/2012] [Indexed: 10/28/2022]
Abstract
ADAM12 (A Disintegrin And Metalloprotease 12), a member of the ADAMs family of transmembrane proteins, is involved in ectodomain shedding, cell-adhesion and signaling, with important implications in cancer. Therefore, mechanisms that regulate the levels and activity of ADAM12 at the cell-surface are possibly crucial in these contexts. We here investigated internalization and subsequent recycling or degradation of ADAM12 as a potentially important regulatory mechanism. Our results show that ADAM12 is constitutively internalized primarily via the clathrin-dependent pathway and is subsequently detected in both early and recycling endosomes. The protease activity of ADAM12 does not influence this internalization mechanism. Analysis of essential elements for internalization established that proline-rich regions in the cytoplasmic domain of ADAM12, previously shown to interact with Src-homology 3 domains, were necessary for proper internalization. These sites in the ADAM12 cytoplasmic domain interacted with the adaptor protein growth factor receptor-bound protein 2 (Grb2) and knockdown of Grb2 markedly reduced ADAM12 internalization. These studies establish that internalization is indeed a mechanism that regulates ADAM cell surface levels and show that ADAM12 internalization involves the clathrin-dependent pathway and Grb2.
Collapse
Affiliation(s)
- Dorte Stautz
- Department of Biomedical Sciences & Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
21
|
Kim YM, Kim J, Heo SC, Shin SH, Do EK, Suh DS, Kim KH, Yoon MS, Lee TG, Kim JH. Proteomic identification of ADAM12 as a regulator for TGF-β1-induced differentiation of human mesenchymal stem cells to smooth muscle cells. PLoS One 2012; 7:e40820. [PMID: 22808268 PMCID: PMC3396647 DOI: 10.1371/journal.pone.0040820] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 06/13/2012] [Indexed: 12/02/2022] Open
Abstract
Background Transforming growth factor-β1 (TGF-β1) induces the differentiation of human adipose tissue-derived mesenchymal stem cells (hASCs) into smooth muscle cells. Lipid rafts are cholesterol-rich microdomains in cell membranes that reportedly play a key role in receptor-mediated signal transduction and cellular responses. In order to clarify whether lipid rafts are involved in TGF-β1-induced differentiation of hASCs into smooth muscle cells, we analyzed the lipid raft proteome of hASCs. Methods and Results Pretreatment of hASCs with the lipid raft disruptor methyl-β-cyclodextrin abrogated TGF-β1-induced expression of α-smooth muscle actin, a smooth muscle cell marker, suggesting a pivotal role of lipid rafts in TGF-β1-induced differentiation of hASCs to smooth muscle cells. Sucrose density gradient centrifugation along with a shotgun proteomic strategy using liquid chromatography-tandem mass spectrometry identified 1002 individual proteins as the lipid raft proteome, and 242 of these were induced by TGF-β1 treatment. ADAM12, a disintegrin and metalloproteases family member, was identified as the most highly up-regulated protein in response to TGF-β1 treatment. TGF-β1 treatment of hASCs stimulated the production of both ADAM12 protein and mRNA. Silencing of endogenous ADAM12 expression using lentiviral small hairpin RNA or small interfering RNA abrogated the TGF-β1-induced differentiation of hASCs into smooth muscle cells. Conclusions These results suggest a pivotal role for lipid raft-associated ADAM12 in the TGF-β1-induced differentiation of hASCs into smooth muscle cells.
Collapse
Affiliation(s)
- Young Mi Kim
- Medical Research Center for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan, Republic of Korea
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Jaeyoon Kim
- NovaCell Technology Inc., Pohang, Republic of Korea
| | - Soon Chul Heo
- Medical Research Center for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan, Republic of Korea
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Sang Hun Shin
- Medical Research Center for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan, Republic of Korea
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Eun Kyoung Do
- Medical Research Center for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan, Republic of Korea
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Dong-Soo Suh
- Department of Obstetrics and Gynecology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Ki-Hyung Kim
- Department of Obstetrics and Gynecology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Man-Soo Yoon
- Department of Obstetrics and Gynecology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | | | - Jae Ho Kim
- Medical Research Center for Ischemic Tissue Regeneration, School of Medicine, Pusan National University, Yangsan, Republic of Korea
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
- * E-mail:
| |
Collapse
|
22
|
Leyme A, Bourd-Boittin K, Bonnier D, Falconer A, Arlot-Bonnemains Y, Théret N. Identification of ILK as a new partner of the ADAM12 disintegrin and metalloprotease in cell adhesion and survival. Mol Biol Cell 2012; 23:3461-72. [PMID: 22767580 PMCID: PMC3431925 DOI: 10.1091/mbc.e11-11-0918] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
ILK is identified as a new partner for ADAM12L cell signaling functions. ADAM12L colocalizes with ILK at focal adhesions and induces the Akt-dependent survival pathway via stimulation of β1 integrins and activation of PI3K. This effect is independent of ADAM12L proteolytic activity and involves its cytoplasmic domain. Based on its shedding and binding activities, the disintegrin and metalloprotease 12 (ADAM12) has been implicated in cell signaling. Here we investigate the intracellular protein interaction network of the transmembrane ADAM12L variant using an integrative approach. We identify the integrin-linked kinase (ILK) as a new partner for ADAM12L cellular functions. We demonstrate that ADAM12L coimmunoprecipitates with ILK in cells and that its cytoplasmic tail is required for this interaction. In human cultured hepatic stellate cells (HSCs), which express high levels of endogenous ADAM12L and ILK, the two proteins are redistributed to focal adhesions upon stimulation of a β1 integrin–dependent pathway. We show that down-regulation of ADAM12L in HSCs leads to cytoskeletal disorganization and loss of adhesion. Conversely, up-regulation of ADAM12L induces the Akt Ser-473 phosphorylation-dependent survival pathway via stimulation of β1 integrins and activation of phosphoinositide 3-kinase (PI3K). Depletion of ILK inhibits this effect, which is independent of ADAM12L proteolytic activity and involves its cytoplasmic domain. We further demonstrate that overexpression of ADAM12L promotes kinase activity from ILK immunoprecipitates. Our data suggest a new role for ADAM12L in mediating the functional association of ILK with β1 integrin to regulate cell adhesion/survival through a PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Anthony Leyme
- Institut National de la Santé et de la Recherche Médicale, UMR1085, Institut de Recherche en Santé, Environnement et Travail, Université de Rennes 1, 35043 Rennes, France
| | | | | | | | | | | |
Collapse
|
23
|
Stautz D, Wewer UM, Kveiborg M. Functional analysis of a breast cancer-associated mutation in the intracellular domain of the metalloprotease ADAM12. PLoS One 2012; 7:e37628. [PMID: 22662180 PMCID: PMC3360752 DOI: 10.1371/journal.pone.0037628] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 04/26/2012] [Indexed: 11/18/2022] Open
Abstract
A recently identified breast cancer-associated mutation in the metalloprotease ADAM12 alters a potential dileucine trafficking signal, which could affect protein processing and cellular localization. ADAM12 belongs to the group of A Disintegrin And Metalloproteases (ADAMs), which are typically membrane-associated proteins involved in ectodomain shedding, cell-adhesion, and signaling. ADAM12 as well as several members of the ADAM family are over-expressed in various cancers, correlating with disease stage. Three breast cancer-associated somatic mutations were previously identified in ADAM12, and two of these, one in the metalloprotease domain and another in the disintegrin domain, were investigated and found to result in protein misfolding, retention in the secretory pathway, and failure of zymogen maturation. The third mutation, p.L792F in the ADAM12 cytoplasmic tail, was not investigated, but is potentially significant given its location within a di-leucine motif, which is recognized as a potential cellular trafficking signal. The present study was motivated both by the potential relevance of this documented mutation to cancer, as well as for determining the role of the di-leucine motif in ADAM12 trafficking. Expression of ADAM12 p.L792F in mammalian cells demonstrated quantitatively similar expression levels and zymogen maturation as wild-type (WT) ADAM12, as well as comparable cellular localizations. A cell surface biotinylation assay demonstrated that cell surface levels of ADAM12 WT and ADAM12 p.L792F were similar and that internalization of the mutant occurred at the same rate and extent as for ADAM12 WT. Moreover, functional analysis revealed no differences in cell proliferation or ectodomain shedding of epidermal growth factor (EGF), a known ADAM12 substrate between WT and mutant ADAM12. These data suggest that the ADAM12 p.L792F mutation is unlikely to be a driver (cancer causing)-mutation in breast cancer.
Collapse
Affiliation(s)
| | | | - Marie Kveiborg
- Department of Biomedical Sciences & Biotech Research and Innovation Centre, Copenhagen University, Ole Maaløes Vej, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
24
|
Fried D, Böhm BB, Krause K, Burkhardt H. ADAM15 protein amplifies focal adhesion kinase phosphorylation under genotoxic stress conditions. J Biol Chem 2012; 287:21214-23. [PMID: 22544741 DOI: 10.1074/jbc.m112.347120] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ADAM15, a disintegrin and metalloproteinase, is capable of counteracting genotoxic stress-induced apoptosis by the suppression of caspase-3 activation. A cell line expressing the membrane-bound ADAM15 without its cytoplasmic tail, however, lost this anti-apoptotic property, suggesting a crucial role of the intracellular domain as a scaffold for recruitment of survival signal-transducing kinases. Accordingly, an enhanced phosphorylation of FAK at Tyr-397, Tyr-576, and Tyr-861 was detected upon genotoxic stress by camptothecin in ADAM15-transfected T/C28a4 cells, but not in transfectants expressing an ADAM15 mutant without the cytoplasmic tail. Accordingly, a specific binding of the cytoplasmic ADAM15 domain to the C terminus of FAK could be shown by mammalian two-hybrid, pulldown, and far Western studies. In cells expressing full-length ADAM15, a concomitant activation of Src at Tyr-416 was detected upon camptothecin exposure. Cells transfected with a chimeric construct consisting of the extracellular IL-2 receptor α-chain and the cytoplasmic ADAM15 domain were IL-2-stimulated to prove that the ADAM15 tail can transduce a percepted extracellular signal to enhance FAK and Src phosphorylation. Our studies further demonstrate Src binding to FAK but not a direct Src interaction with ADAM15, suggesting FAK as a critical intracellular adaptor for ADAM15-dependent enhancement of FAK/Src activation. Moreover, the apoptosis induction elicited by specific inhibitors (PP2, FAK 14 inhibitor) of FAK/Src signaling was significantly reduced by ADAM15 expression. The newly uncovered counter-regulatory response to genotoxic stress in a chondrocytic survival pathway is potentially also relevant to apoptosis resistance in neoplastic growth.
Collapse
Affiliation(s)
- Dorothee Fried
- Division of Rheumatology, Goethe University Hospital Frankfurt am Main, 60590 Frankfurt am Main, Germany
| | | | | | | |
Collapse
|
25
|
Molecular analysis of melatonin-induced changes in breast cancer cells: microarray study of anti-cancer effect of melatonin. BIOCHIP JOURNAL 2011. [DOI: 10.1007/s13206-011-5409-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
26
|
Linder S, Wiesner C, Himmel M. Degrading devices: invadosomes in proteolytic cell invasion. Annu Rev Cell Dev Biol 2011; 27:185-211. [PMID: 21801014 DOI: 10.1146/annurev-cellbio-092910-154216] [Citation(s) in RCA: 290] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Podosomes and invadopodia, collectively known as invadosomes, are cell-matrix contacts in a variety of cell types, such as monocytic cells or cancer cells, that have to cross tissue barriers. Both structures share an actin-rich core, which distinguishes them from other matrix contacts, and are regulated by a multitude of signaling pathways including RhoGTPases, kinases, actin-associated proteins, and microtubule-dependent transport. Invadosomes recruit and secrete proteinases and are thus able to lyse extracellular matrix components. They are therefore considered to be potential key structures in proteolytic cell invasion in both physiological and pathological settings. This review provides an overview of the field, with special focus on current developments such as intracellular transport processes, ultrastructural analysis, the possible involvement of invadosomes in disease, and the tentative identification of invadosomes in 3D environments and in vivo.
Collapse
Affiliation(s)
- Stefan Linder
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, 20246 Hamburg, Germany.
| | | | | |
Collapse
|
27
|
Cousin H, Abbruzzese G, Kerdavid E, Gaultier A, Alfandari D. Translocation of the cytoplasmic domain of ADAM13 to the nucleus is essential for Calpain8-a expression and cranial neural crest cell migration. Dev Cell 2011; 20:256-63. [PMID: 21316592 DOI: 10.1016/j.devcel.2010.12.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Revised: 11/01/2010] [Accepted: 12/06/2010] [Indexed: 12/24/2022]
Abstract
ADAMs are transmembrane metalloproteases that control cell behavior by cleaving both cell adhesion and signaling molecules. The cytoplasmic domain of ADAMs can regulate the proteolytic activity by controlling the subcellular localization and/or the activation of the protease domain. Here, we show that the cytoplasmic domain of ADAM13 is cleaved and translocates into the nucleus. Preventing this translocation renders the protein incapable of promoting cranial neural crest (CNC) cell migration in vivo, without affecting its proteolytic activity. In addition, the cytoplasmic domain of ADAM13 regulates the expression of multiple genes in CNC, including the protease Calpain8-a. Restoring the expression of Calpain8-a is sufficient to rescue CNC migration in the absence of the ADAM13 cytoplasmic domain. This study shows that the cytoplasmic domain of ADAM metalloproteases can perform essential functions in the nucleus of cells and may contribute substantially to the overall function of the protein.
Collapse
Affiliation(s)
- Hélène Cousin
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003, USA.
| | | | | | | | | |
Collapse
|
28
|
McLachlan S, Lee SM, Steele TM, Hawthorne PL, Zapala MA, Eskin E, Schork NJ, Anderson GJ, Vulpe CD. In silico QTL mapping of basal liver iron levels in inbred mouse strains. Physiol Genomics 2010; 43:136-47. [PMID: 21062905 DOI: 10.1152/physiolgenomics.00025.2010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Both iron deficiency and iron excess are detrimental in many organisms, and previous studies in both mice and humans suggest that genetic variation may influence iron status in mammals. However, these genetic factors are not well defined. To address this issue, we measured basal liver iron levels in 18 inbred strains of mice of both sexes on a defined iron diet and found ∼4-fold variation in liver iron in males (lowest 153 μg/g, highest 661 μg/g) and ∼3-fold variation in females (lowest 222 μg/g, highest 658 μg/g). We carried out a genome-wide association mapping to identify haplotypes underlying differences in liver iron and three other related traits (copper and zinc liver levels, and plasma diferric transferrin levels) in a subset of 14 inbred strains for which genotype information was available. We identified two putative quantitative trait loci (QTL) that contain genes with a known role in iron metabolism: Eif2ak1 and Igf2r. We also identified four putative QTL that reside in previously identified iron-related QTL and 22 novel putative QTL. The most promising putative QTL include a 0.22 Mb region on Chromosome 7 and a 0.32 Mb region on Chromosome 11 that both contain only one candidate gene, Adam12 and Gria1, respectively. Identified putative QTL are good candidates for further refinement and subsequent functional studies.
Collapse
Affiliation(s)
- Stela McLachlan
- Department of Nutritional Science and Toxicology, University of California Berkeley, Berkeley, CA 94720, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Albrechtsen R, Stautz D, Sanjay A, Kveiborg M, Wewer UM. Extracellular engagement of ADAM12 induces clusters of invadopodia with localized ectodomain shedding activity. Exp Cell Res 2010; 317:195-209. [PMID: 20951132 DOI: 10.1016/j.yexcr.2010.10.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 09/27/2010] [Accepted: 10/01/2010] [Indexed: 12/23/2022]
Abstract
Invadopodia are dynamic actin structures at the cell surface that degrade extracellular matrix and act as sites of signal transduction. The biogenesis of invadopodia, including the mechanisms regulating their formation, composition, and turnover is not entirely understood. Here, we demonstrate that antibody ligation of ADAM12, a transmembrane disintegrin and metalloprotease, resulted in the rapid accumulation of invadopodia with extracellular matrix-degrading capacity in epithelial cells expressing the αvβ3 integrin and active c-Src kinase. The induction of invadopodia clusters required an intact c-Src interaction site in the ADAM12 cytoplasmic domain, but was independent of the catalytic activity of ADAM12. Caveolin-1 and transmembrane protease MMP14/MT1-MMP were both present in the ADAM12-induced clusters of invadopodia, and cholesterol depletion prevented their formation, suggesting that lipid-raft microdomains are involved in the process. Importantly, our data demonstrate that ADAM12-mediated ectodomain shedding of epidermal growth factor receptor ligands can occur within these invadopodia. Such localized growth factor signalling offers an interesting novel biological concept highly relevant to the properties of carcinoma cells, which often show upregulated ADAM12 and β3 integrin expression, together with high levels of c-Src kinase activity.
Collapse
Affiliation(s)
- Reidar Albrechtsen
- Department of Biomedical Sciences & Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
30
|
Selective inhibition of ADAM12 catalytic activity through engineering of tissue inhibitor of metalloproteinase 2 (TIMP-2). Biochem J 2010; 430:79-86. [PMID: 20533908 DOI: 10.1042/bj20100649] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The disintegrin and metalloprotease ADAM12 has important functions in normal physiology as well as in diseases, such as cancer. Little is known about how ADAM12 confers its pro-tumorigenic effect; however, its proteolytic capacity is probably a key component. Thus selective inhibition of ADAM12 activity may be of great value therapeutically and as an investigative tool to elucidate its mechanisms of action. We have previously reported the inhibitory profile of TIMPs (tissue inhibitor of metalloproteinases) against ADAM12, demonstrating in addition to TIMP-3, a unique ADAM-inhibitory activity of TIMP-2. These findings strongly suggest that it is feasible to design a TIMP mutant selectively inhibiting ADAM12. With this purpose, we characterized the molecular determinants of the ADAM12-TIMP complex formation as compared with known molecular requirements for TIMP-mediated inhibition of ADAM17/TACE (tumour necrosis factor alpha-converting enzyme). Kinetic analysis using a fluorescent peptide substrate demonstrated that the molecular interactions of N-TIMPs (N-terminal domains of TIMPs) with ADAM12 and TACE are for the most part comparable, yet revealed strikingly unique features of TIMP-mediated ADAM12 inhibition. Intriguingly, we found that removal of the AB-loop in N-TIMP-2, which is known to impair its interaction with TACE, resulted in increased affinity to ADAM12. Importantly, using a cell-based epidermal growth factor-shedding assay, we demonstrated for the first time an inhibitory activity of TIMPs against the transmembrane ADAM12-L (full-length ADAM12), verifying the distinctive inhibitory abilities of N-TIMP-2 and engineered N-TIMP-2 mutants in a cellular environment. Taken together, our findings support the idea that a distinctive ADAM12 inhibitor with future therapeutic potential can be designed.
Collapse
|
31
|
Hernández I, Moreno JL, Zandueta C, Montuenga L, Lecanda F. Novel alternatively spliced ADAM8 isoforms contribute to the aggressive bone metastatic phenotype of lung cancer. Oncogene 2010; 29:3758-69. [PMID: 20453887 DOI: 10.1038/onc.2010.130] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
ADAMs (a disintegrin and metalloprotease) are transmembrane proteins involved in a variety of physiological processes and tumorigenesis. Recently, ADAM8 has been associated with poor prognosis of lung cancer. However, its contribution to tumorigenesis in the context of lung cancer metastasis remains unknown. Native ADAM8 expression levels were lower in lung cancer cell lines. In contrast, we identified and characterized two novel spliced isoforms encoding truncated proteins, Delta18a and Delta14', which were present in several tumor cell lines and not in normal cells. Overexpression of Delta18a protein resulted in enhanced invasive activity in vitro. ADAM8 and its Delta14' isoform expression levels were markedly increased in lung cancer cells, in conditions mimicking tumor microenvironment. Moreover, addition of supernatants from Delta14'-overexpressing cells resulted in a significant increase in tartrate-resistant acid phosphatase+ cells in osteoclast cultures in vitro. These findings were associated with increased pro-osteoclastogenic cytokines interleukin (IL)-8 and IL-6 protein levels. Furthermore, lung cancer cells overexpressing Delta14' increased prometastatic activity with a high tumor burden and increased osteolysis in a murine model of bone metastasis. Thus, the expression of truncated forms of ADAM8 by the lung cancer cells may result in the specific upregulation of their invasive and osteoclastogenic activities in the bone microenvironment. These findings suggest a novel mechanism of tumor-induced osteolysis in metastatic bone colonization.
Collapse
Affiliation(s)
- I Hernández
- Adhesion and Metastasis Laboratory, Division of Oncology, Center for Applied Biomedical Research (CIMA), University of Navarra, Pamplona, Spain
| | | | | | | | | |
Collapse
|
32
|
Abstract
Kinases and proteases are responsible for two fundamental regulatory mechanisms--phosphorylation and proteolysis--that orchestrate the rhythms of life and death in all organisms. Recent studies have highlighted the elaborate interplay between both post-translational regulatory systems. Many intracellular or pericellular proteases are regulated by phosphorylation, whereas multiple kinases are activated or inactivated by proteolytic cleavage. The functional consequences of this regulatory crosstalk are especially relevant in the different stages of cancer progression. What are the clinical implications derived from the fertile dialogue between kinases and proteases in cancer?
Collapse
Affiliation(s)
- Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología, Universidad de Oviedo, 33006 Oviedo, Spain.
| | | |
Collapse
|