1
|
Borkúti P, Kristó I, Szabó A, Kovács Z, Vilmos P. FERM domain-containing proteins are active components of the cell nucleus. Life Sci Alliance 2024; 7:e202302489. [PMID: 38296350 PMCID: PMC10830384 DOI: 10.26508/lsa.202302489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 02/05/2024] Open
Abstract
The FERM domain is a conserved and widespread protein module that appeared in the common ancestor of amoebae, fungi, and animals, and is therefore now found in a wide variety of species. The primary function of the FERM domain is localizing to the plasma membrane through binding lipids and proteins of the membrane; thus, for a long time, FERM domain-containing proteins (FDCPs) were considered exclusively cytoskeletal. Although their role in the cytoplasm has been extensively studied, the recent discovery of the presence and importance of cytoskeletal proteins in the nucleus suggests that FDCPs might also play an important role in nuclear function. In this review, we collected data on their nuclear localization, transport, and possible functions, which are still scattered throughout the literature, with special regard to the role of the FERM domain in these processes. With this, we would like to draw attention to the exciting, new dimension of the role of FDCPs, their nuclear activity, which could be an interesting novel direction for future research.
Collapse
Affiliation(s)
| | | | - Anikó Szabó
- HUN-REN Biological Research Centre, Szeged, Hungary
| | - Zoltán Kovács
- HUN-REN Biological Research Centre, Szeged, Hungary
- Doctoral School of Multidisciplinary Medical Science, University of Szeged, Szeged, Hungary
| | - Péter Vilmos
- HUN-REN Biological Research Centre, Szeged, Hungary
| |
Collapse
|
2
|
Duan B, Qin Z, Gu X, Li Y. Migfilin: Cell Adhesion Effect and Comorbidities. Onco Targets Ther 2022; 15:411-422. [PMID: 35469339 PMCID: PMC9034862 DOI: 10.2147/ott.s357355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/04/2022] [Indexed: 11/28/2022] Open
Abstract
Cell adhesion manifests as cell linkages to neighboring cells and/or the extracellular matrix (ECM). Migfilin is a widely expressed adhesion protein. It comprises three LIM domains in the C-terminal region and one proline-rich sequence in the N-terminal region. Through interplay with its various binding partners, such as Kindlin-2, Filamin, vasodilator-stimulated phosphoprotein (VASP) protein and the transcription factor CSX, Migfilin facilitates the dynamic association of connecting actomyosin fibers, orchestrating cell morphogenetic movement and cell adhesion, proliferation, migration, invasion, differentiation and signal transduction. In this review, to further elucidate the functional contributions of and pathogenesis induced by Migfilin, we focused on the structure of Migfilin and the targets which it directly binds with. We also summarized the role of Migfilin and its binding partners in the progression of different diseases and malignancies. As a possible candidate for coordinating various cellular processes and because of its association with both the pathogenesis and progression of certain tumors, Migfilin likely has utility as a therapeutic target against multiple diseases in the clinic.
Collapse
Affiliation(s)
- Baoyu Duan
- Department of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, People’s Republic of China
| | - Ziyao Qin
- Department of Research and Development, Shanghai Institute of Biological Products Co., Ltd., Shanghai, People’s Republic of China
| | - Xuefeng Gu
- Department of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, People’s Republic of China
- Xuefeng Gu, Department of Pharmacy, 279 Zhouzhu Road, Shanghai, 201318, People’s Republic of China, Tel +86 21 6588 3180, Email
| | - Yanfei Li
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, People’s Republic of China
- Correspondence: Yanfei Li, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, 1500 Zhouyuan Road, Shanghai, 201318, People’s Republic of China, Tel +86 21 6588 3180 Email
| |
Collapse
|
3
|
Godbout E, Son DO, Hume S, Boo S, Sarrazy V, Clément S, Kapus A, Wehrle-Haller B, Bruckner-Tuderman L, Has C, Hinz B. Kindlin-2 Mediates Mechanical Activation of Cardiac Myofibroblasts. Cells 2020; 9:cells9122702. [PMID: 33348602 PMCID: PMC7766948 DOI: 10.3390/cells9122702] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
We identify the focal adhesion protein kindlin-2 as player in a novel mechanotransduction pathway that controls profibrotic cardiac fibroblast to myofibroblast activation. Kindlin-2 is co-upregulated with the myofibroblast marker α-smooth muscle actin (α-SMA) in fibrotic rat hearts and in human cardiac fibroblasts exposed to fibrosis-stiff culture substrates and pro-fibrotic TGF-β1. Stressing fibroblasts using ferromagnetic microbeads, stretchable silicone membranes, and cell contraction agonists all result in kindlin-2 translocation to the nucleus. Overexpression of full-length kindlin-2 but not of kindlin-2 missing a putative nuclear localization sequence (∆NLS kindlin-2) results in increased α-SMA promoter activity. Downregulating kindlin-2 with siRNA leads to decreased myofibroblast contraction and reduced α-SMA expression, which is dependent on CC(A/T)-rich GG(CArG) box elements in the α-SMA promoter. Lost myofibroblast features under kindlin-2 knockdown are rescued with wild-type but not ∆NLS kindlin-2, indicating that myofibroblast control by kindlin-2 requires its nuclear translocation. Because kindlin-2 can act as a mechanotransducer regulating the transcription of α-SMA, it is a potential target to interfere with myofibroblast activation in tissue fibrosis.
Collapse
Affiliation(s)
- Elena Godbout
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada; (E.G.); (D.O.S.); (S.H.); (S.B.); (V.S.)
| | - Dong Ok Son
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada; (E.G.); (D.O.S.); (S.H.); (S.B.); (V.S.)
| | - Stephanie Hume
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada; (E.G.); (D.O.S.); (S.H.); (S.B.); (V.S.)
| | - Stellar Boo
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada; (E.G.); (D.O.S.); (S.H.); (S.B.); (V.S.)
| | - Vincent Sarrazy
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada; (E.G.); (D.O.S.); (S.H.); (S.B.); (V.S.)
| | - Sophie Clément
- Division of Clinical Pathology, University Hospital, University of Geneva School of Medicine, 1211 Geneva 4, Switzerland;
| | - Andras Kapus
- Keenan Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada;
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universitaire, University of Geneva, 1211 Geneva 4, Switzerland;
| | - Leena Bruckner-Tuderman
- Medical Center and Medical Faculty, University of Freiburg, 79104 Freiburg, Germany; (L.B.-T.); (C.H.)
| | - Cristina Has
- Medical Center and Medical Faculty, University of Freiburg, 79104 Freiburg, Germany; (L.B.-T.); (C.H.)
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada; (E.G.); (D.O.S.); (S.H.); (S.B.); (V.S.)
- Correspondence: ; Tel.: +1-416-978-8728
| |
Collapse
|
4
|
Singh Z. Leiomyosarcoma: A rare soft tissue cancer arising from multiple organs. JOURNAL OF CANCER RESEARCH AND PRACTICE 2018. [DOI: 10.1016/j.jcrpr.2017.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
5
|
Rognoni E, Ruppert R, Fässler R. The kindlin family: functions, signaling properties and implications for human disease. J Cell Sci 2016; 129:17-27. [PMID: 26729028 DOI: 10.1242/jcs.161190] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The kindlin (or fermitin) family of proteins comprises three members (kindlin-1,-2 and -3) of evolutionarily conserved focal adhesion (FA) proteins, whose best-known task is to increase integrin affinity for a ligand (also referred as integrin activation) through binding of β-integrin tails. The consequence of kindlin-mediated integrin activation and integrin-ligand binding is cell adhesion, spreading and migration, assembly of the extracellular matrix (ECM), cell survival, proliferation and differentiation. Another hallmark of kindlins is their involvement in disease. Mutations in the KINDLIN-1 (also known as FERMT1) gene cause Kindler syndrome (KS)--in which mainly skin and intestine are affected, whereas mutations in the KINDLIN-3 (also known as FERMT3) gene cause leukocyte adhesion deficiency type III (LAD III), which is characterized by impaired extravasation of blood effector cells and severe, spontaneous bleedings. Also, aberrant expression of kindlins in various forms of cancer and in tissue fibrosis has been reported. Although the malfunctioning of integrins represent a major cause leading to kindlin-associated diseases, increasing evidence also point to integrin-independent functions of kindlins that play an important role in the pathogenesis of certain disease aspects. Furthermore, isoform-specific kindlin functions have been discovered, explaining, for example, why loss of kindlins differentially affects tissue stem cell homeostasis or tumor development. This Commentary focuses on new and isoform-specific kindlin functions in different tissues and discusses their potential role in disease development and progression.
Collapse
Affiliation(s)
- Emanuel Rognoni
- Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Raphael Ruppert
- Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Reinhard Fässler
- Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| |
Collapse
|
6
|
Yan M, Zhang L, Wu Y, Gao L, Yang W, Li J, Chen Y, Jin X. Increased expression of kindlin-2 is correlated with hematogenous metastasis and poor prognosis in patients with clear cell renal cell carcinoma. FEBS Open Bio 2016; 6:660-5. [PMID: 27398306 PMCID: PMC4932446 DOI: 10.1002/2211-5463.12063] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 04/04/2016] [Accepted: 04/05/2016] [Indexed: 12/23/2022] Open
Abstract
Kindlin‐2 is involved in activating the integrin signaling pathway which plays an important role in regulating cancer cell invasion. However, the role of kindlin‐2 may vary among cancer types. The aim of this study was to explore the possible association between kindlin‐2 and clear cell renal cell carcinoma (ccRCC), and its potential role in the prognosis of ccRCC. Immunohistochemistry assays were used to examine kindlin‐2 expression levels in cancer tissues obtained from 336 patients with ccRCC. The correlation between kindlin‐2 expression levels and pathologic variables was then analyzed. In addition, the association between kindlin‐2 expression levels and survival time was analyzed by Kaplan–Meier survival curves and log‐rank tests. Of 336 ccRCC patients, 199 had high levels of kindlin‐2 expression, while 137 had low kindlin‐2 expression levels. Patients at a late stage of ccRCC (stage III or IV) were more likely to have high kindlin‐2 expression levels than those at an early stage (stage I or II) (χ2 = 4.72, P = 0.03). Patients with high levels of kindlin‐2 expression had higher risk of hematogenous metastasis (χ2 = 6.70, P = 0.01) than those with low levels of kindlin‐2 expression. In addition, the survival time was significantly shorter for patients with high levels of kindlin‐2 expression than for those with low levels of kindlin‐2 expression (P = 0.001 for overall survival [OS] and P = 0.002 for disease‐free survival [DFS]). Multivariate survival analysis based on the Cox proportional hazards model showed that high kindlin‐2 expression levels had a hazard risk (HR) of 1.76 for OS (95% CI 1.19–2.62, P = 0.005) and an HR of 1.47 for DFS (95% CI = 1.05–2.06, P = 0.026). By comparison, lymph node metastasis had an HR of 1.48 for OS (95% CI 1.04–2.10, P = 0.029) and an HR of 1.41 for DFS (95% CI 1.04–1.93, P = 0.029). This study provided strong evidence that increased kindlin‐2 expression might be involved in promoting tumor invasiveness and leading to a poor prognosis of ccRCC.
Collapse
Affiliation(s)
- Meisi Yan
- Department of Pathology Harbin Medical University China
| | - Lei Zhang
- Department of Pathology Harbin Medical University China
| | - Yiqi Wu
- Department of Pathology Harbin Medical University China
| | - Lei Gao
- Electron Microscopy Centre Harbin Medical University China
| | - Weiwei Yang
- Department of Pathology Harbin Medical University China
| | - Jing Li
- Electron Microscopy Centre Harbin Medical University China
| | - Yubing Chen
- Department of Radiotherapy Second Hospital of Jilin University Changchun China
| | - Xiaoming Jin
- Department of Pathology Harbin Medical University China
| |
Collapse
|
7
|
Cao HH, Zhang SY, Shen JH, Wu ZY, Wu JY, Wang SH, Li EM, Xu LY. A three-protein signature and clinical outcome in esophageal squamous cell carcinoma. Oncotarget 2015; 6:5435-48. [PMID: 25605255 PMCID: PMC4467159 DOI: 10.18632/oncotarget.3102] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/28/2014] [Indexed: 02/05/2023] Open
Abstract
Current staging is inadequate to precisely predict clinical outcome of esophageal squamous cell carcinoma (ESCC) and determine treatment choices, which vary from operation alone to intensive multimodal regimens. The purpose of this study is to investigate the prognostic values of an immunohistochemistry-based three-protein signature model in patients with ESCC. We determined the protein expression of Annexin II, cofilin 1, ezrin, fascin, kindlin-2, moesin, MTSS1, myosin-9, profilin-1, Rac1, radixin, ROCK2, talin, tensin and villin 1 in a test cohort including 110 formalin-fixed, paraffin-embedded esophageal curative resection specimens by tissue microarrays (TMAs). A three-protein signature elicited from the protein cluster, Annexin II, kindlin-2, and myosin-9, was validated by TMAs on an independent cohort of 147 specimens. The expression of three-protein signature was highly predictive of ESCC overall survival (OS) and disease-free survival (DFS) in both generation and validation datasets. Regression analysis shows that this three-protein signature is an independent predictor for OS and DFS. Furthermore, the predictive ability of these 3 biomarkers in combination is more robust than that of each individual biomarker. This study demonstrates a clinically applicable prognostic model that accurately predicts ESCC patient survival and/or tumor recurrence, and thus could serve as a complement to current risk stratification approaches.
Collapse
Affiliation(s)
- Hui-Hui Cao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, P.R. China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, P.R. China
- Departments of Pathology, Zhuhai People's Hospital, Zhuhai, Guangdong, P.R. China
| | - Shi-Yi Zhang
- Departments of Oncology Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong, P.R. China
| | - Jin-Hui Shen
- Departments of Pathology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong, P.R. China
| | - Zhi-Yong Wu
- Departments of Oncology Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong, P.R. China
| | - Jian-Yi Wu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, P.R. China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, P.R. China
| | - Shao-Hong Wang
- Departments of Pathology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong, P.R. China
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, P.R. China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, P.R. China
| | - Li-Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, P.R. China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, P.R. China
| |
Collapse
|
8
|
Ge YS, Liu D, Jia WD, Li JS, Ma JL, Yu JH, Xu GL. Kindlin-2: a novel prognostic biomarker for patients with hepatocellular carcinoma. Pathol Res Pract 2014; 211:198-202. [PMID: 25618552 DOI: 10.1016/j.prp.2014.09.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 05/31/2014] [Accepted: 09/12/2014] [Indexed: 01/12/2023]
Abstract
The association of aberrant expression of Kindlin-2 with tumor progression has been reported in recent years. The purpose of this study was to investigate the expression of Kindlin-2 in hepatocellular carcinoma (HCC), and to evaluate its clinical and prognostic significance. The mRNA and protein levels of Kindlin-2 in HCC and adjacent non-cancerous tissues were examined by real-time PCR and western blotting. The relationships between Kindlin-2 expression, clinicopathological features and postoperative survival of HCC patients were also evaluated. Kindlin-2 expression was higher in HCC tissues as compared to adjacent non-cancerous tissues at both mRNA and protein levels (P<0.05, respectively). Positive expression of Kindlin-2 was significantly correlated with larger tumor size (P=0.034), capsular invasion (P=0.009), microvascular invasion (P=0.028) and poor prognosis of HCC patients (P<0.001). Moreover, multivariate survival analysis identified Kindlin-2 as an independent prognostic factor for overall and disease-free survival of HCC patients (P=0.018 and 0.001, respectively). Taken together, our findings suggested that Kindlin-2 was highly expressed in HCC tissues and was closely related to clinical progression. Therefore, Kindlin-2 protein could be a potential biomarker for predicting poor prognosis of HCC patients after surgery.
Collapse
Affiliation(s)
- Yong-Sheng Ge
- Graduate School of Tianjin Medical University, Tianjin 300070, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Hospital, Hefei 230001, Anhui Province, China; Department of Hepatic Surgery, Anhui Provincial Hospital, Anhui Medical University, Hefei 230001, Anhui Province, China
| | - Dong Liu
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Hospital, Hefei 230001, Anhui Province, China; Department of general surgery, Lu'an People's Hospital, Lu'an 237005, Anhui Province, China
| | - Wei-Dong Jia
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Hospital, Hefei 230001, Anhui Province, China; Department of Hepatic Surgery, Anhui Provincial Hospital, Anhui Medical University, Hefei 230001, Anhui Province, China
| | - Jian-Sheng Li
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Hospital, Hefei 230001, Anhui Province, China; Department of Hepatic Surgery, Anhui Provincial Hospital, Anhui Medical University, Hefei 230001, Anhui Province, China
| | - Jin-Liang Ma
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Hospital, Hefei 230001, Anhui Province, China; Department of Hepatic Surgery, Anhui Provincial Hospital, Anhui Medical University, Hefei 230001, Anhui Province, China
| | - Ji-Hai Yu
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Hospital, Hefei 230001, Anhui Province, China; Department of Hepatic Surgery, Anhui Provincial Hospital, Anhui Medical University, Hefei 230001, Anhui Province, China
| | - Ge-Liang Xu
- Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Hospital, Hefei 230001, Anhui Province, China; Department of Hepatic Surgery, Anhui Provincial Hospital, Anhui Medical University, Hefei 230001, Anhui Province, China.
| |
Collapse
|
9
|
Kindlin-2 inhibits serous epithelial ovarian cancer peritoneal dissemination and predicts patient outcomes. Biochem Biophys Res Commun 2014; 446:187-94. [DOI: 10.1016/j.bbrc.2014.02.087] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 02/19/2014] [Indexed: 12/29/2022]
|
10
|
Yu Y, Wu J, Guan L, Qi L, Tang Y, Ma B, Zhan J, Wang Y, Fang W, Zhang H. Kindlin 2 promotes breast cancer invasion via epigenetic silencing of the microRNA200 gene family. Int J Cancer 2013; 133:1368-79. [PMID: 23483548 DOI: 10.1002/ijc.28151] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 02/20/2013] [Indexed: 02/06/2023]
Abstract
Kindlin 2, as a focal adhesion protein, controls integrin activation and regulates Wnt signaling in an integrin-binding independent manner. However, the association of Kindlin 2 with cancer-related microRNAs is unknown. Here, we report that Kindlin 2 markedly downregulates the expression of miR-200 family by inducing CpG island hypermethylation. Mechanistically, Kindlin 2 forms a complex with DNMT3A in the cell nucleus and the two proteins co-occupy the promoter of miRNA-200b. Functionally, repression of miR-200b is required for Kindlin 2-induced breast cancer cell invasion and tumor formation. Our data indicate that Kindlin 2 plays a novel role in epigenetic repression of miR-200 family, a mechanism that promotes breast cancer invasion.
Collapse
Affiliation(s)
- Yu Yu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education of China, Peking University Health Science Center, Beijing, 100191, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Zhao T, Guan L, Yu Y, Pei X, Zhan J, Han L, Tang Y, Li F, Fang W, Zhang H. Kindlin-2 promotes genome instability in breast cancer cells. Cancer Lett 2013; 330:208-16. [DOI: 10.1016/j.canlet.2012.11.043] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 11/26/2012] [Accepted: 11/27/2012] [Indexed: 12/31/2022]
|
12
|
A feedback regulation between Kindlin-2 and GLI1 in prostate cancer cells. FEBS Lett 2013; 587:631-8. [PMID: 23337877 DOI: 10.1016/j.febslet.2012.12.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 12/05/2012] [Accepted: 12/21/2012] [Indexed: 01/15/2023]
Abstract
Kindlin-2 is engaged in tumor progression. However, the mechanism accounting for Kindlin-2 regulation in tumor cells remained largely unknown. Here, we report a regulatory loop between Kindlin-2 and GLI1, an effector of Hedgehog signaling pathway. We show that Kindlin-2 is transcriptionally downregulated via GLI1 occupancy on the Kindlin-2 promoter. Adversely, we found that Kindlin-2 promotes GLI1 expression through a mechanism involving GSK3β inactivation and is independent of Smoothened. Functionally, knockdown of Kindlin-2 cooperates with cyclopamine, a Smoothened antagonist, to decrease the viability of prostate cancer cells. Taken together, targeting the Kindlin-2-GLI1 feedback loop may facilitate the killing of prostate cancer cells.
Collapse
|
13
|
Shen Z, Ye Y, Kauttu T, Seppänen H, Vainionpää S, Wang S, Mustonen H, Puolakkainen P. The novel focal adhesion gene kindlin-2 promotes the invasion of gastric cancer cells mediated by tumor-associated macrophages. Oncol Rep 2012; 29:791-7. [PMID: 23151599 DOI: 10.3892/or.2012.2137] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 09/20/2012] [Indexed: 12/13/2022] Open
Abstract
Kindlin-2 is a novel focal adhesion gene mediating the cell-extracellular matrix (ECM) adhesion. Tumor-associated macrophages (TAMs) play an important role in linking chronic inflammation to cancer progression. Both kindlin-2 and TAMs have been found to promote the invasion of gastric cancer cells in our previous studies. However, the correlation between kindlin-2 and TAMs remains unclear. Real-time RT-PCR was used to investigate kindlin-2 expression in the AGS, NCI and Hs-746T gastric cancer cell lines co-cultured with TAMs under normal or hypoxic conditions. IL8, IL10, IL11, IL17b, IL18, IL22 and IL24 expressions were measured by real-time RT-PCR in the gastric cancer lines with varying levels of kindlin-2 expression, as well as after downregulation of kindlin-2 mRNA expression by the siRNA method. We found that kindlin-2 was upregulated in all three gastric cancer cell lines when co-cultured with TAMs under normal conditions. Under hypoxic conditions, the induction of kindlin-2 expression induced by macrophages was significantly downregulated in the Hs-746T cell line. IL8, IL11, IL17b, IL22 and IL24 expression was significantly higher in gastric cell lines with high kindlin-2 expression. Downregulation of kindlin-2 mRNA decreased IL10, IL11, IL17b, IL22 and IL24 expression but IL8 and IL18 expression was upregulated. Therefore, the novel focal adhesion gene kindlin-2 may play an important role in promoting the invasion of gastric cancer cells mediated by TAMs through regulating interleukin expression.
Collapse
Affiliation(s)
- Zhanlong Shen
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100044, PR China
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Yu Y, Wu J, Wang Y, Zhao T, Ma B, Liu Y, Fang W, Zhu WG, Zhang H. Kindlin 2 forms a transcriptional complex with β-catenin and TCF4 to enhance Wnt signalling. EMBO Rep 2012; 13:750-8. [PMID: 22699938 DOI: 10.1038/embor.2012.88] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 04/28/2012] [Accepted: 05/21/2012] [Indexed: 11/09/2022] Open
Abstract
Kindlin 2, as a focal adhesion protein, controls integrin activation. However, the association of Kindlin 2 with cancer-related signalling pathways is unknown. Here we identified a new direct interaction between Kindlin 2 and the active β-catenin. Importantly, Kindlin 2 forms a tripartite complex with β-catenin and TCF4. Mechanistically, Kindlin 2 selectively strengthens the occupancy of β-catenin on the Wnt target gene Axin2 and enhances Axin2 gene expression. Functionally, the β-catenin-Axin2-Snail cascade is required for Kindlin 2-induced tumour cell invasion. Our data indicate that Kindlin 2 is a new regulator of Wnt signalling, providing a mechanistic insight into the role of Kindlin 2 in cancer progression.
Collapse
Affiliation(s)
- Yu Yu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, #38 Xue Yuan Road, Beijing 100191, China
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
|
16
|
Gong X, An Z, Wang Y, Guan L, Fang W, Strömblad S, Jiang Y, Zhang H. Kindlin-2 controls sensitivity of prostate cancer cells to cisplatin-induced cell death. Cancer Lett 2010; 299:54-62. [DOI: 10.1016/j.canlet.2010.08.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 08/05/2010] [Accepted: 08/09/2010] [Indexed: 01/06/2023]
|
17
|
Abstract
Focal adhesion kinase (FAK) is a scaffold and tyrosine kinase protein that binds to itself and cellular partners through its four-point-one, ezrin, radixin, moesin (FERM) domain. Recent structural work reveals that regulatory protein partners convert auto-inhibited FAK into its active state by binding to its FERM domain. Further, the identity of FAK FERM domain-interacting proteins yields clues as to how FAK coordinates diverse cellular responses, including cell adhesion, polarization, migration, survival and death, and suggests that FERM domains might mediate information transfer between the cell cortex and nucleus. Importantly, the FAK FERM domain might act as a paradigm for the actions of other FERM domain-containing proteins.
Collapse
|
18
|
An Z, Dobra K, Lock JG, Strömblad S, Hjerpe A, Zhang H. Kindlin-2 is expressed in malignant mesothelioma and is required for tumor cell adhesion and migration. Int J Cancer 2010; 127:1999-2008. [PMID: 20127858 DOI: 10.1002/ijc.25223] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Kindlin-2 is a novel integrin-interacting focal adhesion protein that belongs to the Kindlin family. Focal adhesion proteins control cytoskeleton dynamics and promote cancer cell growth, survival, migration and metastasis. Little is known, however, about expression of Kindlin-2 in association with human cancer. We now reveal high Kindlin-2 expression in malignant mesothelioma (MM) cell lines using an affinity-purified anti-Kindlin-2 antibody. Furthermore, we show by immunohistochemistry that Kindlin-2 is highly expressed in 92 of 102 (90%) MMs with epitheliod; sarcomatoid, biphasic and poorly differentiated morphologies. In addition, Kindlin-2 expression correlates to cell proliferation, suggesting a role for Kindlin-2 in tumor growth. We also detect increased expression of Kindlin-2 at the invasion front of tumors concurrent with increased expression of integrin-linked kinase, a Kindlin-binding protein. Besides the high expression of Kindlin-2 in pleural MMs, pleural metastases of lung adenocarcinoma also express large amounts of Kindlin-2, but not Kindlin-1. Notably, in vitro, when endogenous Kindlin-2 was knocked down with RNAi in MM cells, this impaired cell spreading, adhesion and migration. Overall, our study suggests that heightened expression of Kindlin-2 might contribute to tumor progression in MM.
Collapse
Affiliation(s)
- Zhengwen An
- Unit for Clinical Molecular Biology, Department of Biosciences and Nutrition at Novum, Karolinska Institutet, SE-14183, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
19
|
Affiliation(s)
- Esra Karaköse
- Max Planck Institute of Biochemistry, Department of Molecular Medicine, Am Klopferspitz 18a, Martinsried, 82152, Germany
| | - Herbert B. Schiller
- Max Planck Institute of Biochemistry, Department of Molecular Medicine, Am Klopferspitz 18a, Martinsried, 82152, Germany
| | - Reinhard Fässler
- Max Planck Institute of Biochemistry, Department of Molecular Medicine, Am Klopferspitz 18a, Martinsried, 82152, Germany
| |
Collapse
|
20
|
Bialkowska K, Ma YQ, Bledzka K, Sossey-Alaoui K, Izem L, Zhang X, Malinin N, Qin J, Byzova T, Plow EF. The integrin co-activator Kindlin-3 is expressed and functional in a non-hematopoietic cell, the endothelial cell. J Biol Chem 2010; 285:18640-9. [PMID: 20378539 PMCID: PMC2881789 DOI: 10.1074/jbc.m109.085746] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Revised: 03/16/2010] [Indexed: 01/10/2023] Open
Abstract
Integrin activation is crucial for numerous cellular responses, including cell adhesion, migration, and survival. Recent studies in mice have specifically emphasized the vital role of kindlin-3 in integrin activation. Kindlin-3 deficiency in humans also has now been documented and includes symptoms of bleeding, frequent infections, and osteopetrosis, which are consequences of an inability to activate beta1, beta2, and beta3 integrins. To date, kindlin-3 was thought to be restricted to hematopoietic cells. In this article, we demonstrate that kindlin-3 is present in human endothelial cells derived from various anatomical origins. The mRNA and protein for KINDLIN-3 was detected in endothelial cells by reverse transcription-PCR and Western blots. When subjected to sequencing by mass spectrometry, the protein was identified as authentic kindlin-3 and unequivocally distinguished from KINDLIN-1 and KINDLIN-2 or any other known protein. By quantitative real time PCR, the level of kindlin-3 in endothelial cells was 20-50% of that of kindlin-2. Using knockdown approaches, we show that kindlin-3 plays a role in integrin-mediated adhesion of endothelial cells. This function depends upon the integrin and substrate and is distinct from that of kindlin-2. Formation of tube-like structures in Matrigel also was impaired by kindlin-3 knockdown. Mechanistically, the distinct functions of the kindlins can be traced to differences in their subcellular localization in integrin-containing adhesion structures. Thus, the prevailing view that individual kindlins exert their functions in a cell type-specific manner must now be modified to consider distinct functions of the different family members within the same cell type.
Collapse
Affiliation(s)
- Katarzyna Bialkowska
- From the Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology and
| | - Yan-Qing Ma
- From the Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology and
| | - Kamila Bledzka
- From the Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology and
| | - Khalid Sossey-Alaoui
- From the Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology and
| | - Lahoucine Izem
- the Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Xiaoxia Zhang
- From the Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology and
| | - Nikolay Malinin
- From the Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology and
| | - Jun Qin
- From the Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology and
| | - Tatiana Byzova
- From the Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology and
| | - Edward F. Plow
- From the Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology and
| |
Collapse
|
21
|
Abstract
The Kindlin family of intracellular proteins has recently emerged as key regulators of cellular functions and cell-matrix interactions. The 3 members of this family, Kindlin-1, -2, and -3, perform an essential role in activation of integrin adhesion receptors, and expression of at least 1 Kindlin paralog is required to enable integrin activation in physiologically relevant settings. In humans, deficiencies in Kindlin-3 lead to a number of abnormalities affecting hemostasis, the immune system, and bone function, whereas the lack of Kindlin-1 causes profound skin defects. The importance of Kindlins is underscored by the results of animal knockout studies, which clearly show the indispensable and nonredundant functions of all 3 Kindlins in development and normal physiology. This review discusses recent progress in the studies of Kindlin protein family, emphasizing newly identified functions and potential mechanisms underlying differential activities of the family members.
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW There are three kindlin family members in vertebrates, which have high-sequence homology and a common organization signature with a C-terminal 4.1, ezrin, radixin, moesin (FERM) domain bisected by a pleckstrin-homology domain. Although the cell and tissue distributions of the three kindlins differ, there is a consistent and close interrelationship between kindlins and integrins, and alterations of kindlin expression affect integrin-dependent functions. However, in-vivo data on the functions of the kindlins and their mechanisms of action have been lacking. RECENT FINDINGS Recent studies have shown that deficiencies of each of the three kindlins result in profound and distinct phenotypes, ranging from skin and intestinal defects (kindlin-1), embryonic lethality due to cardiac developmental problems (kindlin-2), to marked abnormalities in platelet, leukocyte and erythrocyte function (kindlin-3). A human disease characterized by bleeding, frequent infections and osteopetrosis has now been attributed to mutations in the gene encoding for kindlin-3. These defects are consistent with recent data showing that kindlins control integrin activation and function. SUMMARY The three members of the kindlin family have now been implicated as essential regulators of integrin function in individual cells and in whole organisms.
Collapse
|
23
|
The Kindlin protein family: new members to the club of focal adhesion proteins. Trends Cell Biol 2009; 19:504-13. [DOI: 10.1016/j.tcb.2009.07.006] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Revised: 07/04/2009] [Accepted: 07/08/2009] [Indexed: 11/19/2022]
|
24
|
How ILK and kindlins cooperate to orchestrate integrin signaling. Curr Opin Cell Biol 2009; 21:670-5. [DOI: 10.1016/j.ceb.2009.05.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Revised: 05/28/2009] [Accepted: 05/29/2009] [Indexed: 12/18/2022]
|
25
|
Shi X, Wu C. A suppressive role of mitogen inducible gene-2 in mesenchymal cancer cell invasion. Mol Cancer Res 2008; 6:715-24. [PMID: 18505917 DOI: 10.1158/1541-7786.mcr-07-2026] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer cell invasion of extracellular matrix (ECM) is essential for dissemination of cancer cells and metastasis. In this study, we have investigated the role of mitogen inducible gene-2 (Mig-2, also known as kindlin-2), a focal adhesion protein whose expression is altered in several types of human cancers, in mesenchymal cancer cell invasion. Mig-2 is abundantly expressed in SK-LMS-1 leiomyosarcoma cells. The level of Mig-2, however, is considerably lower in more invasive HT-1080 fibrosarcoma cells. Overexpression of Mig-2 in HT-1080 and SK-LMS-1 cells substantially reduced their ability to invade ECM in an in vitro Matrigel invasion assay. Conversely, knockdown of Mig-2 markedly increased the invasiveness of these cells. Consistent with a suppressive role in mesenchymal cancer cell invasion, Mig-2 inhibits urokinase-type plasminogen activator (uPA) secretion and pericellular proteolysis. Overexpression of Mig-2 increased uPA accumulation at the intracellular face of cell-ECM adhesions and reduced the level of secreted uPA. Conversely, knockdown of Mig-2 reduced uPA accumulation at the intracellular face of cell-ECM adhesions and increased uPA secretion. Our results reveal an important role of Mig-2 in suppression of mesenchymal cancer cell invasion and shed new light on how altered Mig-2 expression could influence cancer cell invasion.
Collapse
Affiliation(s)
- Xiaohua Shi
- 707B Scaife Hall, Department of Pathology, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15261, USA
| | | |
Collapse
|
26
|
Hayashi T, Shimamura Y, Saegusa T, Horiuchi A, Kobayashi Y, Hiraoka N, Kanai Y, Aburatani H, Sano K, Konishi I. Molecular mechanisms of uterine leiomyosarcomas: involvement of defect in LMP2 expression. GENE REGULATION AND SYSTEMS BIOLOGY 2008; 2:297-305. [PMID: 19787091 PMCID: PMC2733082 DOI: 10.4137/grsb.s470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Patients with uterine leiomyosarcoma (LMS) typically present with vaginal bleeding, pain, and a pelvic mass. Typical presentations with hypercalcemia or eosinophilia have been reported. Radiographic evaluation with combined positron emission tomography/computed tomography may assist in the diagnosis and surveillance of women with uterine LMS. A recently developed risk-assessment index is highly predictive of disease-specific survival. Ovarian preservation does not appear to negatively impact outcome, and the addition of adjuvant therapy after surgical treatment does not seem to improve survival. It is noteworthy that LMP2-deficient mice exhibit spontaneous development of uterine LMS with a disease prevalence of ~37% by 12 months of age. The LMP2 gene is transcribed from a promoter containing an interferon (IFN)-γ-response factor element; thus, the IFN-γ-signal strongly induces LMP2 expression. Furthermore, a recent report demonstrated the loss of ability to induce LMP2 expression, which is an interferon (IFN)-γ-inducible factor, in human uterine LMS tissues and cell lines. Analysis of human uterine LMS shows somatic mutations in the IFNγ signalling pathway, thus the loss of LMP2 induction is attributable to a defect in the earliest steps of the IFN-γ signalling pathway. The discovery of an impaired key cell-signalling pathway may provide new targets for diagnostic approaches and therapeutic intervention.
Collapse
Affiliation(s)
- Takuma Hayashi
- Department of Immunology and Infectious Disease, Shinshu University Graduate School of Medicine, Nagano, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Vasilopoulos Y, Gkretsi V, Armaka M, Aidinis V, Kollias G. Actin cytoskeleton dynamics linked to synovial fibroblast activation as a novel pathogenic principle in TNF-driven arthritis. Ann Rheum Dis 2007; 66 Suppl 3:iii23-8. [PMID: 17934089 PMCID: PMC2095291 DOI: 10.1136/ard.2007.079822] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2007] [Indexed: 12/21/2022]
Abstract
Rheumatoid arthritis is a chronic inflammatory disorder whose origin of defect has been the subject of extensive research during the past few decades. While a number of immune and non-immune cell types participate in the development of chronic destructive inflammation in the arthritic joint, synovial fibroblasts have emerged as key effector cells capable of modulating both joint destruction and propagation of inflammation. Ample evidence of aberrant changes in the morphology and biochemical behaviour of rheumatoid arthritis synovial fibroblasts have established the tissue evading and "transformed" character of this cell type. We have recently demonstrated that actin cytoskeletal rearrangements determine the pathogenic activation of synovial fibroblasts in modelled TNF-mediated arthritis, a finding correlating with similar gene expression changes which we observed in human rheumatoid arthritis synovial fibroblasts. Here, we show that pharmacological inhibition of actin cytoskeleton dynamics alters potential pathogenic properties of the arthritogenic synovial fibroblast, such as proliferation, migration and resistance to apoptosis, indicating novel opportunities for therapeutic intervention in arthritis. Recent advances in this field of research are reviewed and discussed.
Collapse
Affiliation(s)
- Y Vasilopoulos
- Institute of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece
| | | | | | | | | |
Collapse
|
28
|
Papachristou DJ, Gkretsi V, Tu Y, Shi X, Chen K, Larjava H, Rao UNM, Wu C. Increased cytoplasmic level of migfilin is associated with higher grades of human leiomyosarcoma. Histopathology 2007; 51:499-508. [PMID: 17711449 PMCID: PMC2768333 DOI: 10.1111/j.1365-2559.2007.02791.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
AIMS Leiomyosarcomas (LMS) are malignant neoplasms composed of cells that exhibit distinct smooth muscle differentiation. The molecular and cytogenetic features of LMS are complex and no consistent aberrations have been reported to date. Mitogen inducible gene-2 (Mig-2), kindlin and migfilin are recently identified cell-matrix adhesion proteins. The aim was to determine the expression and distribution of these proteins in human smooth muscle tumours of somatic soft tissue. METHODS AND RESULTS Immunohistochemistry was performed on a human LMS tissue microarray and on sections of human leiomyomas (LM) and normal smooth muscle. Migfilin was barely detectable in normal smooth muscle cells, whereas increased levels of migfilin were observed in the majority of LM and LMS. Furthermore, the cytoplasmic level of migfilin was strongly associated with higher tumour grades. Additionally, the cytoplasmic levels of migfilin and Mig-2 were correlated with each other, suggesting an association between the two in the cytoplasm. Kindlin was expressed in normal smooth muscle, LM and LMS, and its level did not correlate with tumour grade. CONCLUSIONS Our results suggest a role for cytoplasmic migfilin in the progression of LMS and identify cytoplasmic migfilin as a potentially important biological marker for human LMS progression.
Collapse
Affiliation(s)
- D J Papachristou
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Gozgit JM, Pentecost BT, Marconi SA, Otis CN, Wu C, Arcaro KF. Use of an aggressive MCF-7 cell line variant, TMX2-28, to study cell invasion in breast cancer. Mol Cancer Res 2007; 4:905-13. [PMID: 17189381 DOI: 10.1158/1541-7786.mcr-06-0147] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
An estrogen receptor-negative variant of the MCF-7 breast cancer cell line, TMX2-28, was used as a model in which to study breast cancer cell invasion. Using a reconstituted basement membrane (Matrigel) assay to evaluate cell invasion, we determined that TMX2-28 cells are more invasive than MCF-7 cells and that the invasiveness of TMX2-28 is similar to that of the aggressive MDA-MB-231 breast cancer cell line. TMX2-28 cells displayed a rounded, epithelial cell-like morphology, suggesting an amoeboid mode of cell invasion, in contrast to the mesenchymal mode of invasion characteristic of spindle-shaped, fibroblast-like MDA-MB-231 cells. Using real-time reverse transcription-PCR, we found that mitogen-inducible gene 2 (MIG2) is expressed at a 17-fold higher level in TMX2-28 cells than in nonaggressive MCF-7 cells and that MIG2 mRNA levels are low in the nontumorigenic human mammary epithelial cell line, 184. We determined that MIG2 plays a role in cell invasion by using small interfering RNA (siRNA) to suppress the expression of MIG2 mRNA levels in TMX2-28 cells. TMX2-28 cell invasion was reduced by 48% when the cells were transfected with siRNAs targeting MIG2, relative to cells transfected with siRNAs against glyceraldehyde-3-phosphate dehydrogenase. Finally, MIG2 expression was evaluated in reductive mammoplasty and breast tumor tissue. Although all 21 normal tissues from reduction mammoplasty showed immunoreactivity for MIG2, ranging from weak (62%) to strong (24%), only half of the 34 formalin-fixed breast tumors showed immunoreactivity for MIG2. Of these 17 positive cases, 10 were considered to overexpress MIG2 (moderate to strong staining). Examination of 30 frozen breast tumors supported the finding that MIG2 is overexpressed in a subset of breast cancers. We suggest that MIG2's normal regulation and function are disrupted in breast cancer.
Collapse
Affiliation(s)
- Joseph M Gozgit
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
30
|
Ussar S, Wang HV, Linder S, Fässler R, Moser M. The Kindlins: subcellular localization and expression during murine development. Exp Cell Res 2006; 312:3142-51. [PMID: 16876785 DOI: 10.1016/j.yexcr.2006.06.030] [Citation(s) in RCA: 199] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Revised: 06/06/2006] [Accepted: 06/13/2006] [Indexed: 12/25/2022]
Abstract
The three Kindlins are a novel family of focal adhesion proteins. The Kindlin-1 (URP1) gene is mutated in Kindler syndrome, the first skin blistering disease affecting actin attachment in basal keratinocytes. Kindlin-2 (Mig-2), the best studied member of this family, binds ILK and Migfilin, which links Kindlin-2 to the actin cytoskeleton. Kindlin-3 is expressed in hematopoietic cells. Here we describe the genomic organization, gene expression and subcellular localization of murine Kindlins-1 to -3. In situ hybridizations showed that Kindlin-1 is preferentially expressed in epithelia, and Kindlin-2 in striated and smooth muscle cells. Kindlins-1 and -2 are both expressed in the epidermis. While both localize to integrin-mediated adhesion sites in cultured keratinocytes Kindlin-2, but not Kindlin-1, colocalizes with E-cadherin to cell-cell contacts in differentiated keratinocytes. Using a Kindlin-3-specific antiserum and an EGFP-tagged Kindlin-3 construct, we could show that Kindlin-3 is present in the F-actin surrounding ring structure of podosomes, which are specialized adhesion structures of hematopoietic cells.
Collapse
Affiliation(s)
- Siegfried Ussar
- Max-Planck-Institute of Biochemistry, Department of Molecular Medicine, D-82152 Martinsried, Germany
| | | | | | | | | |
Collapse
|
31
|
Abstract
The possibility that a fibroid may actually be a smooth muscle tumor of uncertain malignant potential or a leiomyosarcoma often dictates the clinical management of rapidly growing fibroids. This article provides a clinicopathologic background regarding uterine leiomyosarcomas and answers common questions that obstetricians and gynecologists have about the management of rapidly growing fibroids and leiomyosarcoma. The clinician must be aware that the infrequent occurrence of uterine leiomyosarcomas makes it difficult to establish absolutely firm recommendations for the diagnosis and management of this disease, particularly with regard to fertility preservation. Nevertheless, this article addresses major issues that a clinician might face in the evaluation of a smooth muscle tumor of the uterus that clinically may be malignant.
Collapse
Affiliation(s)
- Peter E Schwartz
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA.
| | | |
Collapse
|
32
|
Abstract
Cell-extracellular matrix (ECM) adhesion is crucial for control of cell behavior. It connects the ECM to the intracellular cytoskeleton and transduces bidirectional signals between the extracellular and intracellular compartments. The subcellular machinery that mediates cell-ECM adhesion and signaling is complex. It consists of transmembrane proteins (e.g., integrins) and at least several dozens of membrane-proximal proteins that assemble into a network through multiple protein interactions. Furthermore, despite sharing certain common components, cell-ECM adhesions exhibit considerable heterogeneity in different types of cells (e.g., the cell-ECM adhesions in cardiac myocytes are considerably different from those in fibroblasts). Here, we will first briefly describe the general properties of the integrin-mediated cell-ECM adhesion and signal transduction. Next, we will focus on one of the recently discovered cell-ECM adhesion protein complexes consisting of PINCH, integrin-linked kinase (ILK), and Parvin and use it as an example to illustrate the molecular basis underlying the assembly and functions of cell-ECM adhesions. Finally, we will discuss in detail the structure and regulation of cell-ECM adhesion complexes in cardiac myocytes, which illustrate the importance and complexity of the cell-ECM adhesion structures in organogenesis and diseases.
Collapse
Affiliation(s)
- Jorge L Sepulveda
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | | | | |
Collapse
|