40 results on '"integrin α2"'
Search Results
2. Overexpressed integrin alpha 2 inhibits the activation of the transforming growth factor β pathway in pancreatic cancer via the TFCP2-SMAD2 axis
- Author
-
Hongkun Cai, Feng Guo, Shuang Wen, Xin Jin, Heshui Wu, and Dianyun Ren
- Subjects
Pancreatic cancer ,Integrin α2 ,Transcription factor CP2 ,SMAD2 ,TGF-β ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Integrin alpha 2 (ITGA2) has been recently reported to be an oncogene and to play crucial roles in tumor cell proliferation, invasion, metastasis, and angiogenesis. Our previous study showed that ITGA2 was overexpressed in pancreatic cancer and promoted its progression. However, the mechanism of ITGA2 overexpression and other mechanisms for promoting the progression of pancreatic cancer are still unclear. Methods The GEPIA database was used to confirm the expression of ITGA2 in pancreatic cancer. To verify the influence of ITGA2 and TGF-β on the morphological changes of pancreatic cancer and tumor cell progression, we conduct CCK8 test, plate cloning, flow cytometry experiments and animal experiments. Then we conduct Western blot, RT-qPCR to explore the relationship between ITGA2 and TGF-β, and then find the key molecules which can regulate them by immunoprecipitation, Western blot, RT-qPCR, CHIP, nuclear and cytoplasmic separation test. Results The results of the present study show that the abnormal activation of KRAS induced the overexpression of ITGA2 in pancreatic cancer. Moreover, ITGA2 expression significantly suppressed the activation of the TGF-β pathway. ITGA2 silencing enhanced the anti-pancreatic cancer proliferation and tumor growth effects of TGF-β. Mechanistically, ITGA2 expression suppressed the activation of the TGF-β pathway by inhibiting the SMAD2 expression transcriptionally. In addition, it interacted with and inhibited the nuclear translocation of TFCP2, which induced the SMAD2 expression as a transcription factor. Furthermore, TFCP2 also induced ITGA2 expression as a transcription factor, and the TFCP2 feedback regulated the ITGA2-TFCP2-SMAD2 pathway. Conclusions Taken together, these results indicated that ITGA2 expression could inhibit the activation of the TGF-β signaling pathway in pancreatic cancer via the TFCP2-SMAD2 axis. Therefore, ITGA2, by effectively enhancing the anti-cancer effects of TGF- β, might be a potential clinical therapeutic target for pancreatic cancer.
- Published
- 2022
- Full Text
- View/download PDF
3. Integrin α2 in the microenvironment and the tumor compartment of digestive (gastrointestinal) cancers: emerging regulators and therapeutic opportunities.
- Author
-
Liu T, Gu Y, Zhang Y, and Li Y
- Abstract
Integrins are a family of cell surface membrane receptors and play a crucial role in facilitating bidirectional cell signaling. Integrin α2 (ITGA2) is expressed across a range of cell types, including epithelial cells, platelets, megakaryocytes, and fibroblasts, where it functions as a surface marker and it is implicated in the cell movements. The most recent findings have indicated that ITAG2 has the potential to function as a novel regulatory factor in cancer, responsible for driving tumorigenesis, inducing chemoresistance, regulating genomic instability and remodeling tumor microenvironment. Hence, we primarily focus on elucidating the biological function and mechanism of ITGA2 within the digestive tumor microenvironment, while highlighting its prospective utilization as a therapeutic target for cancer therapy., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Liu, Gu, Zhang and Li.)
- Published
- 2024
- Full Text
- View/download PDF
4. The small-molecule protein ligand interface stabiliser E7820 induces differential cell line specific responses of integrin α2 expression
- Author
-
Michael David Hülskamp, Daniel Kronenberg, and Richard Stange
- Subjects
E7820 ,Integrin α2 ,CAPERα ,SPLINTS ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The mechanism of small-molecule stabilised protein-protein interactions is of growing interest in the pharmacological discovery process. A plethora of different substances including the aromatic sulphonamide E7820 have been identified to act by such a mechanism. The process of E7820 induced CAPERα degradation and the resultant transcriptional down regulation of integrin α2 expression has previously been described for a variety of different cell lines and been made responsible for E7820’s antiangiogenic activity. Currently the application of E7820 in the treatment of various malignancies including pancreas carcinoma and breast cancer is being investigated in pre-clinical and clinical trials. It has been shown, that integrin α2 deficiency has beneficial effects on bone homeostasis in mice. To transfer E7820 treatment to bone-related pathologies, as non-healing fractures, osteoporosis and bone cancer might therefore be beneficial. However, at present no data is available on the effect of E7820 on osseous cells or skeletal malignancies. Methods Pre-osteoblastic (MC3T3 and Saos-2) cells and endothelial (eEnd2 cells and HUVECs) cells, each of human and murine origin respectively, were investigated. Vitality assay with different concentrations of E7820 were performed. All consecutive experiments were done at a final concentration of 50 ng/ml E7820. The expression and production of integrin α2 and CAPERα were investigated by quantitative real-time PCR and western blotting. Expression of CAPERα splice forms was differentiated by semi-quantitiative reverse transcriptase PCR. Results Here we present the first data showing that E7820 can increase integrin α2 expression in the pre-osteoblast MC3T3 cell line whilst also reproducing canonical E7820 activity in HUVECs. We show that the aberrant activity of E7820 in MC3T3 cells is likely due to differential activity of CAPERα at the integrin α2 promoter, rather than due to differential CAPERα degradation or differential expression of CAPERα spliceforms. Conclusion The results presented here indicate that E7820 may not be suitable to treat certain malignancies of musculoskeletal origin, due to the increase in integrin α2 expression it may induce. Further investigation of the differential functioning of CAPERα and the integrin α2 promoter in cells of various origin would however be necessary to more clearly differentiate between cell lines that will positively respond to E7820 from those that will not.
- Published
- 2021
- Full Text
- View/download PDF
5. Overexpressed integrin alpha 2 inhibits the activation of the transforming growth factor β pathway in pancreatic cancer via the TFCP2-SMAD2 axis.
- Author
-
Cai, Hongkun, Guo, Feng, Wen, Shuang, Jin, Xin, Wu, Heshui, and Ren, Dianyun
- Subjects
- *
PANCREATIC cancer , *INTEGRINS , *PANCREATIC tumors , *TRANSCRIPTION factors , *SMAD proteins - Abstract
Background: Integrin alpha 2 (ITGA2) has been recently reported to be an oncogene and to play crucial roles in tumor cell proliferation, invasion, metastasis, and angiogenesis. Our previous study showed that ITGA2 was overexpressed in pancreatic cancer and promoted its progression. However, the mechanism of ITGA2 overexpression and other mechanisms for promoting the progression of pancreatic cancer are still unclear. Methods: The GEPIA database was used to confirm the expression of ITGA2 in pancreatic cancer. To verify the influence of ITGA2 and TGF-β on the morphological changes of pancreatic cancer and tumor cell progression, we conduct CCK8 test, plate cloning, flow cytometry experiments and animal experiments. Then we conduct Western blot, RT-qPCR to explore the relationship between ITGA2 and TGF-β, and then find the key molecules which can regulate them by immunoprecipitation, Western blot, RT-qPCR, CHIP, nuclear and cytoplasmic separation test. Results: The results of the present study show that the abnormal activation of KRAS induced the overexpression of ITGA2 in pancreatic cancer. Moreover, ITGA2 expression significantly suppressed the activation of the TGF-β pathway. ITGA2 silencing enhanced the anti-pancreatic cancer proliferation and tumor growth effects of TGF-β. Mechanistically, ITGA2 expression suppressed the activation of the TGF-β pathway by inhibiting the SMAD2 expression transcriptionally. In addition, it interacted with and inhibited the nuclear translocation of TFCP2, which induced the SMAD2 expression as a transcription factor. Furthermore, TFCP2 also induced ITGA2 expression as a transcription factor, and the TFCP2 feedback regulated the ITGA2-TFCP2-SMAD2 pathway. Conclusions: Taken together, these results indicated that ITGA2 expression could inhibit the activation of the TGF-β signaling pathway in pancreatic cancer via the TFCP2-SMAD2 axis. Therefore, ITGA2, by effectively enhancing the anti-cancer effects of TGF- β, might be a potential clinical therapeutic target for pancreatic cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
6. Overexpressed ITGA2 promotes malignant tumor aggression by up-regulating PD-L1 expression through the activation of the STAT3 signaling pathway
- Author
-
Dianyun Ren, Jingyuan Zhao, Yan Sun, Dan Li, Zibo Meng, Bo Wang, Ping Fan, Zhiqiang Liu, Xin Jin, and Heshui Wu
- Subjects
Integrin α2 ,Programmed death-ligand 1 ,Phosphorylation of STAT3 ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Recent studies have reported that Integrin alpha 2 (ITGA2) plays an essential role in tumor cell proliferation, invasion, metastasis, and angiogenesis. An abnormally expressed ITGA2 correlates with unfavorable prognoses in multiple types of cancer. However, the specific mechanism of how ITGA2 contributes to tumorigenesis remains unclear. Methods The GEPIA web tool was used to find the clinical relevance of ITGA2 in cancer, and this significance was verified using Western blotting analysis of paired patient tissues and immunohistochemistry of the pancreatic cancer tissue. Functional assays, such as the MTS assay, colony formation assay, and transwell assay, were used to determine the biological role of ITGA2 in human cancer. The relationship between ITGA2 and programmed death-ligand 1 (PD-L1) was examined using Western blot analysis, RT-qPCR assay, and immunohistochemistry. The protein-protein interaction between ITGA2 and STAT3 was detected via co-immunoprecipitation. Results Our study showed that ITGA2 was markedly overexpressed in several malignant tumor cells and clinical tissues. Blocking ITGA2 inhibited the proliferation and invasion ability of cancer cells significantly, whereas overexpressed ITGA2 increased the degree of those processes considerably. Additionally, the RNA-seq assay indicated that ITGA2 transcriptionally regulated the expression of PD-L1 in pancreatic cancer. We also demonstrated that ITGA2 interacted with STAT3 and up-regulated the phosphorylation of STAT3; this interaction might involve the mechanism of ITGA2 inducing PD-L1 expression in cancer cells. Our results suggest that ITGA2 plays a critical role in cancer cell progression and the regulation of PD-L1 by activating the STAT3 pathway. Conclusions We identified a novel mechanism by which ITGA2 plays a critical role in modulating cancer immune response by transcriptionally increasing the expression of PD-L1 in cancer cells. Thus, targeting ITGA2 is an effective method to enhance the efficacy of checkpoint immunotherapy against cancer.
- Published
- 2019
- Full Text
- View/download PDF
7. The small-molecule protein ligand interface stabiliser E7820 induces differential cell line specific responses of integrin α2 expression.
- Author
-
Hülskamp, Michael David, Kronenberg, Daniel, and Stange, Richard
- Subjects
- *
INTEGRINS , *CELL lines , *BONE cells , *REVERSE transcriptase , *BREAST cancer - Abstract
Background: The mechanism of small-molecule stabilised protein-protein interactions is of growing interest in the pharmacological discovery process. A plethora of different substances including the aromatic sulphonamide E7820 have been identified to act by such a mechanism. The process of E7820 induced CAPERα degradation and the resultant transcriptional down regulation of integrin α2 expression has previously been described for a variety of different cell lines and been made responsible for E7820's antiangiogenic activity. Currently the application of E7820 in the treatment of various malignancies including pancreas carcinoma and breast cancer is being investigated in pre-clinical and clinical trials. It has been shown, that integrin α2 deficiency has beneficial effects on bone homeostasis in mice. To transfer E7820 treatment to bone-related pathologies, as non-healing fractures, osteoporosis and bone cancer might therefore be beneficial. However, at present no data is available on the effect of E7820 on osseous cells or skeletal malignancies.Methods: Pre-osteoblastic (MC3T3 and Saos-2) cells and endothelial (eEnd2 cells and HUVECs) cells, each of human and murine origin respectively, were investigated. Vitality assay with different concentrations of E7820 were performed. All consecutive experiments were done at a final concentration of 50 ng/ml E7820. The expression and production of integrin α2 and CAPERα were investigated by quantitative real-time PCR and western blotting. Expression of CAPERα splice forms was differentiated by semi-quantitiative reverse transcriptase PCR.Results: Here we present the first data showing that E7820 can increase integrin α2 expression in the pre-osteoblast MC3T3 cell line whilst also reproducing canonical E7820 activity in HUVECs. We show that the aberrant activity of E7820 in MC3T3 cells is likely due to differential activity of CAPERα at the integrin α2 promoter, rather than due to differential CAPERα degradation or differential expression of CAPERα spliceforms.Conclusion: The results presented here indicate that E7820 may not be suitable to treat certain malignancies of musculoskeletal origin, due to the increase in integrin α2 expression it may induce. Further investigation of the differential functioning of CAPERα and the integrin α2 promoter in cells of various origin would however be necessary to more clearly differentiate between cell lines that will positively respond to E7820 from those that will not. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
8. Hierarchical Micro-Nano Topography Promotes Cell Adhesion and Osteogenic Differentiation via Integrin α2-PI3K-AKT Signaling Axis
- Author
-
Huimin Zheng, Yujuan Tian, Qian Gao, Yingjie Yu, Xianyou Xia, Zhipeng Feng, Feng Dong, Xudong Wu, and Lei Sui
- Subjects
topography ,adhesion ,osteogenic differentiation ,integrin α2 ,PI3K-AKT ,Biotechnology ,TP248.13-248.65 - Abstract
Surface topography dictates important aspects of cell biological behaviors. In our study, hierarchical micro-nano topography (SLM-AHT) with micro-scale grooves and nano-scale pores was fabricated and compared with smooth topography (S) and irregular micro-scale topography (SLA) surfaces to investigate mechanism involved in cell-surface interactions. Integrin α2 had a higher expression level on SLM-AHT surface compared with S and SLA surfaces, and the expression levels of osteogenic markers icluding Runx2, Col1a1, and Ocn were concomitantly upregulated on SLM-AHT surface. Moreover, formation of mature focal adhesions were significantly enhanced in SLM-AHT group. Noticablely, silencing integrin α2 could wipe out the difference of osteogenic gene expression among surfaces with different topography, indicating a crucial role of integrin α2 in topography induced osteogenic differentiation. In addition, PI3K-AKT signaling was proved to be regulated by integrin α2 and consequently participate in this process. Taken together, our findings illustrated that integrin α2-PI3K-AKT signaling axis plays a key role in hierarchical micro-nano topography promoting cell adhesion and osteogenic differentiation.
- Published
- 2020
- Full Text
- View/download PDF
9. Expression dynamics of integrin α2, α3, and αV upon osteogenic differentiation of human mesenchymal stem cells.
- Author
-
Lee, Hyun Min, Seo, Se-Ri, Kim, Jeeseung, Kim, Min Kyu, Seo, Hyosun, Kim, Kyoung Soo, Jang, Young-Joo, and Ryu, Chun Jeih
- Subjects
- *
MESENCHYMAL stem cell differentiation , *INTEGRINS , *ADIPOGENESIS , *DENTAL pulp , *PERIODONTAL ligament - Abstract
Background: The differentiation of human mesenchymal stem cells (hMSCs) into osteoblasts (OBs) is a prerequisite for bone formation. However, little is known about the definitive surface markers for OBs during osteogenesis. Methods: To study the surface markers on OBs, we generated and used monoclonal antibodies (MAbs) against surface molecules on transforming growth factor-β1 (TGF-β1)-treated cancer cells. The generated MAbs were further selected toward expression changes on hMSCs cultured with TGF-β1/bone morphogenetic protein-2 (BMP-2) or osteogenic differentiation medium (ODM) by flow cytometry. Immunoprecipitation and mass spectrometry were performed to identify target antigens of selected MAbs. Expression changes of the target antigens were evaluated in hMSCs, human periodontal ligament cells (hPDLCs), and human dental pulp cells (hDPCs) during osteogenic and adipogenic differentiation by quantitative polymerase chain reaction (qPCR) and flow cytometry. hMSCs were also sorted by the MAbs using magnetic-activated cell sorting system, and osteogenic potential of sorted cells was evaluated via Alizarin Red S (ARS) staining and qPCR. Results: The binding reactivity of MR14-E5, one of the MAbs, was downregulated in hMSCs with ODM while the binding reactivity of ER7-A7, ER7-A8, and MR1-B1 MAbs was upregulated. Mass spectrometry and overexpression identified that MR14-E5, ER7-A7/ER7-A8, and MR1-B1 recognized integrin α2, α3, and αV, respectively. Upon osteogenic differentiation of hMSCs, the expression of integrin α2 was drastically downregulated, but the expression of integrin α3 and αV was upregulated in accordance with upregulation of osteogenic markers. Expression of integrin α3 and αV was also upregulated in hPDLCs and hDPCs during osteogenic differentiation. Cell sorting showed that integrin αV-high hMSCs have a greater osteogenic potential than integrin αV-low hMSCs upon the osteogenic differentiation of hMSCs. Cell sorting further revealed that the surface expression of integrin αV is more dramatically induced even in integrin αV-low hMSCs. Conclusion: These findings suggest that integrin α3 and αV induction is a good indicator of OB differentiation. These findings also shed insight into the expression dynamics of integrins upon osteogenic differentiation of hMSCs and provide the reason why different integrin ligands are required for OB differentiation of hMSCs. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
10. The small-molecule protein ligand interface stabiliser E7820 induces differential cell line specific responses of integrin α2 expression
- Author
-
Hülskamp, Michael David, Kronenberg, Daniel, Stange, Richard, and Universitäts- und Landesbibliothek Münster
- Subjects
Indoles ,610 Medicine and health ,SPLINTS ,Integrin alpha2 ,Ligands ,Cell Line ,Mice ,Human Umbilical Vein Endothelial Cells ,Animals ,Humans ,Protein Isoforms ,ddc:610 ,Promoter Regions, Genetic ,CAPERα ,RC254-282 ,Sulfonamides ,Osteoblasts ,Research ,RNA-Binding Proteins ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Integrin α2 ,Up-Regulation ,E7820 ,Proteolysis ,Medicine and health ,Trans-Activators - Abstract
Background: The mechanism of small-molecule stabilised protein-protein interactions is of growing interest in the pharmacological discovery process. A plethora of different substances including the aromatic sulphonamide E7820 have been identified to act by such a mechanism. The process of E7820 induced CAPERα degradation and the resultant transcriptional down regulation of integrin α2 expression has previously been described for a variety of different cell lines and been made responsible for E7820’s antiangiogenic activity. Currently the application of E7820 in the treatment of various malignancies including pancreas carcinoma and breast cancer is being investigated in pre-clinical and clinical trials. It has been shown, that integrin α2 deficiency has beneficial effects on bone homeostasis in mice. To transfer E7820 treatment to bone-related pathologies, as non-healing fractures, osteoporosis and bone cancer might therefore be beneficial. However, at present no data is available on the effect of E7820 on osseous cells or skeletal malignancies. Methods: Pre-osteoblastic (MC3T3 and Saos-2) cells and endothelial (eEnd2 cells and HUVECs) cells, each of human and murine origin respectively, were investigated. Vitality assay with different concentrations of E7820 were performed. All consecutive experiments were done at a final concentration of 50 ng/ml E7820. The expression and production of integrin α2 and CAPERα were investigated by quantitative real-time PCR and western blotting. Expression of CAPERα splice forms was differentiated by semi-quantitiative reverse transcriptase PCR. Results: Here we present the first data showing that E7820 can increase integrin α2 expression in the preosteoblast MC3T3 cell line whilst also reproducing canonical E7820 activity in HUVECs. We show that the aberrant activity of E7820 in MC3T3 cells is likely due to differential activity of CAPERα at the integrin α2 promoter, rather than due to differential CAPERα degradation or differential expression of CAPERα spliceforms. Conclusion: The results presented here indicate that E7820 may not be suitable to treat certain malignancies of musculoskeletal origin, due to the increase in integrin α2 expression it may induce. Further investigation of the differential functioning of CAPERα and the integrin α2 promoter in cells of various origin would however be necessary to more clearly differentiate between cell lines that will positively respond to E7820 from those that will not., Finanziert über die DEAL-Vereinbarung mit Wiley 2019-2022.
- Published
- 2023
11. Collagen Assembly at the Cell Surface: Dogmas Revisited
- Author
-
Moses Musiime, Joan Chang, Uwe Hansen, Karl E. Kadler, Cédric Zeltz, and Donald Gullberg
- Subjects
collagen fibrillogenesis ,fibronectin ,integrin ,integrin α11 ,integrin α2 ,integrin α5 ,Cytology ,QH573-671 - Abstract
With the increased awareness about the importance of the composition, organization, and stiffness of the extracellular matrix (ECM) for tissue homeostasis, there is a renewed need to understand the details of how cells recognize, assemble and remodel the ECM during dynamic tissue reorganization events. Fibronectin (FN) and fibrillar collagens are major proteins in the ECM of interstitial matrices. Whereas FN is abundant in cell culture studies, it is often only transiently expressed in the acute phase of wound healing and tissue regeneration, by contrast fibrillar collagens form a persistent robust scaffold in healing and regenerating tissues. Historically fibrillar collagens in interstitial matrices were seen merely as structural building blocks. Cell anchorage to the collagen matrix was thought to be indirect and occurring via proteins like FN and cell surface-mediated collagen fibrillogenesis was believed to require a FN matrix. The isolation of four collagen-binding integrins have challenged this dogma, and we now know that cells anchor directly to monomeric forms of fibrillar collagens via the α1β1, α2β1, α10β1 and α11β1 integrins. The binding of these integrins to the mature fibrous collagen matrices is more controversial and depends on availability of integrin-binding sites. With increased awareness about the importance of characterizing the total integrin repertoire on cells, including the integrin collagen receptors, the idea of an absolute dependence on FN for cell-mediated collagen fibrillogenesis needs to be re-evaluated. We will summarize data suggesting that collagen-binding integrins in vitro and in vivo are perfectly well suited for nucleating and supporting collagen fibrillogenesis, independent of FN.
- Published
- 2021
- Full Text
- View/download PDF
12. Influence of collagen‐based integrin α1 and α2 mediated signaling on human mesenchymal stem cell osteogenesis in three dimensional contexts.
- Author
-
Becerra‐Bayona, Silvia M., Guiza‐Arguello, Viviana R., Russell, Brooke, Höök, Magnus, and Hahn, Mariah S.
- Abstract
Collagen I interactions with integrins α1 and α2 are known to support human mesenchymal stem cell (hMSC) osteogenesis. Nonetheless, elucidating the relative impact of specific integrin interactions has proven challenging, in part due to the complexity of native collagen. In the present work, we employed two collagen‐mimetic proteins—Scl2‐2 and Scl2‐3— to compare the osteogenic effects of integrin α1 versus α2 signaling. Scl2‐2 and Scl2‐3 were both derived from Scl2‐1, a triple helical protein lacking known cell adhesion, cytokine binding, and matrix metalloproteinase sites. However, Scl2‐2 and Scl2‐3 were each engineered to display distinct collagen‐based cell adhesion motifs: GFPGER (binding integrins α1 and α2) or GFPGEN (binding only integrin α1), respectively. hMSCs were cultured within poly(ethylene glycol) (PEG) hydrogels containing either Scl2‐2 or Scl2‐3 for 2 weeks. PEG‐Scl2‐2 gels were associated with increased hMSC osterix expression, osteopontin production, and calcium deposition relative to PEG‐Scl2‐3 gels. These data indicate that integrin α2 signaling may have an increased osteogenic effect relative to integrin α1. Since p38 is activated by integrin α2 but not by integrin α1, hMSCs were further cultured in PEG‐Scl2‐2 hydrogels in the presence of a p38 inhibitor. Results suggest that p38 activity may play a key role in collagen‐supported hMSC osteogenesis. This knowledge can be used toward the rational design of scaffolds which intrinsically promote hMSC osteogenesis. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 2594–2604, 2018. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
13. Integrin α2 marks a niche of trophoblast progenitor cells in first trimester human placenta.
- Author
-
Lee, Cheryl Q. E., Turco, Margherita Y., Gardner, Lucy, Simons, Benjamin D., Hemberger, Myriam, and Moffett, Ashley
- Subjects
- *
PREGNANCY , *TROPHOBLAST , *PLACENTA - Abstract
During pregnancy the trophoblast cells of the placenta are the only fetal cells in direct contact with maternal blood and decidua. Their functions include transport of nutrients and oxygen, secretion of pregnancy hormones, remodelling of the uterine arteries, and communicating with maternal cells. Despite the importance of trophoblast cells in placental development and successful pregnancy, little is known about the identity, location and differentiation of human trophoblast progenitors. We identify a proliferative trophoblast niche at the base of the cytotrophoblast cell columns in first trimester placentas that is characterised by integrin α2 (ITGA2) expression. Pulse-chase experiments with 5-iodo-2...-deoxyuridine indicate that these cells might contribute to both villous (VCT) and extravillous (EVT) lineages. These proliferating trophoblast cells can be isolated by flow cytometry using ITGA2 as a marker and express genes from both VCT and EVT. Microarray expression analysis shows that ITAG2+ cells display a unique transcriptional signature, including genes involved in NOTCH signalling, and exhibit a combination of epithelial and mesenchymal characteristics. ITGA2 thus marks a niche allowing the study of pure populations of trophoblast progenitor cells. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
14. Polymorphism in Integrin ITGA2 is Associated with Ischemic Stroke and Altered Serum Cholesterol in Chinese Individuals
- Author
-
Jian-Xia Lu, Zhong-Qian Lu, Wan-Xiang Wang, Shao-lan Zhang, Juan Zhi, and Zheng-Ping Chen
- Subjects
Integrin α2 ,Integrin β3 ,ischemic stroke ,Gene polymorphism ,plasma lipid ,Medicine - Abstract
Background: Recent studies have reported contrasting results regarding the association of polymorphisms in two integrin genes, ITGA2 and ITGB3, with ischemic stroke. Aims: The present study aimed to investigate the correlation between the ITGA2 C807T and ITGB3 T176C polymorphic loci with ischemic stroke, as well as plasma lipid and lipoprotein levels. Study Design: Case control study. Methods: Human venous blood samples were collected from patients admitted for ischemic stroke (n=350, ‘patients’) and healthy individuals (n=300, ‘controls’). Blood was genotyped at these loci by polymerase chain reaction-restriction fragment length polymorphism. Plasma lipid and lipoprotein levels were measured by routine enzymatic, masking, and turbidimetry methods. Results: As expected, total cholesterol, triglycerides, and low-density lipoprotein were all significantly higher in patients than in controls (p
- Published
- 2014
- Full Text
- View/download PDF
15. Targeting integrin α2 as potential strategy for radiochemosensitization of glioblastoma.
- Author
-
Korovina I, Vehlow A, Temme A, and Cordes N
- Subjects
- Humans, Animals, Mice, Integrin alpha2 therapeutic use, Drug Resistance, Neoplasm, Temozolomide therapeutic use, Cell Line, Tumor, Xenograft Model Antitumor Assays, Antineoplastic Agents, Alkylating therapeutic use, Glioblastoma pathology, Brain Neoplasms pathology
- Abstract
Background: Glioblastoma (GBM) is a fast-growing primary brain tumor characterized by high invasiveness and resistance. This results in poor patient survival. Resistance is caused by many factors, including cell-extracellular matrix (ECM) interactions. Here, we addressed the role of adhesion protein integrin α2, which we identified in a high-throughput screen for novel potential targets in GBM cells treated with standard therapy consisting of temozolomide (TMZ) and radiation., Methods: In our study, we used a range of primary/stem-like and established GBM cell models in vitro and in vivo. To identify regulatory mechanisms, we employed high-throughput kinome profiling, Western blotting, immunofluorescence staining, reporter, and activity assays., Results: Our data showed that integrin α2 is overexpressed in GBM compared to normal brain and, that its deletion causes radiochemosensitization. Similarly, invasion and adhesion were significantly reduced in TMZ-irradiated GBM cell models. Furthermore, we found that integrin α2-knockdown impairs the proliferation of GBM cells without affecting DNA damage repair. At the mechanistic level, we found that integrin α2 affects the activity of activating transcription factor 1 (ATF1) and modulates the expression of extracellular signal-regulated kinase 1 (ERK1) regulated by extracellular signals. Finally, we demonstrated that integrin α2-deficiency inhibits tumor growth and thereby prolongs the survival of mice with orthotopically growing GBM xenografts., Conclusions: Taken together our data suggest that integrin α2 may be a promising target to overcome GBM resistance to radio- and chemotherapy. Thus, it would be worth evaluating how efficient and safe the adjuvant use of integrin α2 inhibitors is to standard radio(chemo)therapy in GBM., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
16. Regulation and functions of integrin α2 in cell adhesion and disease
- Author
-
Valery Adorno-Cruz and Huiping Liu
- Subjects
0301 basic medicine ,lcsh:QH426-470 ,Angiogenesis ,Protein subunit ,Integrin ,Biochemistry ,Article ,CD49b ,03 medical and health sciences ,0302 clinical medicine ,Molecular Biology ,Genetics (clinical) ,lcsh:R5-920 ,biology ,Chemistry ,Cell adhesion molecule ,Cell growth ,CD29 ,Molecular mechanisms ,Cell Biology ,Integrin α2 ,Signaling ,3. Good health ,Cell biology ,lcsh:Genetics ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,lcsh:Medicine (General) ,Regulation - Abstract
Integrins are cell adhesion molecules that are composed of an alpha (α) subunit and a beta (β) subunit with affinity for different extracellular membrane components. The integrin family includes 24 known members that actively regulate cellular growth, differentiation, and apoptosis. Each integrin heterodimer has a particular function in defined contexts as well as some partially overlapping features with other members in the family. As many reviews have covered the general integrin family in molecular and cellular studies in life science, this review will focus on the specific regulation, function, and signaling of integrin α2 subunit (CD49b, VLA-2; encoded by the gene ITGA2) in partnership with β1 (CD29) subunit in normal and cancer cells. Its roles in cell adhesion, cell motility, angiogenesis, stemness, and immune/blood cell regulations are discussed. The pivotal role of integrin α2 in many diseases such as cancer suggests its potential to be used as a novel therapeutic target. Keywords: Integrin α2, CD49b, Molecular mechanisms, Regulation, Signaling
- Published
- 2019
17. Altered oxidative status and integrin expression in cyclosporine A-treated oral epithelial cells.
- Author
-
Sardarian, Ahmadreza, Andisheh Tadbir, Azadeh, Zal, Fatemeh, Amini, Fatemeh, Jafarian, Aida, Khademi, Fatemeh, and Mostafavi-Pour, Zohreh
- Subjects
- *
OXIDANT status , *INTEGRINS , *GENE expression , *CYCLOSPORINE , *EPITHELIAL cells , *TRANSPLANTATION of organs, tissues, etc. , *PATIENTS - Abstract
Background and objective: Cyclosporine A (CsA) is an immunosuppressive agent administered to transplant patients. A well-known reported oral side effect of CsA consumption is gingival overgrowth (GO). Changes in the expression of integrins occurring in the gingiva following CsA treatment have been reported but these reports are mainly concerned with the connective tissue of the gingiva. In this study we targeted the alterations in the oral epithelium using KB cells, an oral epithelial cell line. Methods: Cultured oral epithelial cells were treated with increasing concentrations of CsA (0.1, 1 and 10 µg/mL) and the molecular changes involving antioxidant enzymes [glutathione peroxidase (GPx) and glutathione reductase (GR)] and the level of reactive oxygen species (ROS) were measured. Quantitative real-time PCR was used to assess the expression of selected integrins (α2, α5 and β1). Results: At CsA concentration above 0.1 µg/mL GPx demonstrated an increase in activity while GR activity and the level of reduced glutathione were diminished ( p < 0.05). α5 and β1 integrin were downregulated at all treatment concentrations of CsA while α2 integrin presented this effect at concentrations above 1 µg/mL ( p < 0.05). Conclusion: The results suggest a possible role for oxidative stress and the altered expression of integrins in the pathology of CsA-induced gingival overgrowth. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
18. Overexpressed ITGA2 promotes malignant tumor aggression by up-regulating PD-L1 expression through the activation of the STAT3 signaling pathway
- Author
-
Ren, Dianyun, Zhao, Jingyuan, Sun, Yan, Li, Dan, Meng, Zibo, Wang, Bo, Fan, Ping, Liu, Zhiqiang, Jin, Xin, and Wu, Heshui
- Published
- 2019
- Full Text
- View/download PDF
19. Induction of integrin α2 in a highly bone metastatic human prostate cancer cell line: roles of RANKL and AR under three-dimensional suspension culture.
- Author
-
Ziaee, Shabnam and Chung, Leland W. K.
- Subjects
- *
INTEGRINS , *PROSTATE cancer , *BONE metastasis , *CELL lines , *CELL growth , *CELL adhesion - Abstract
Background: Prostate cancer (PCa) bone metastasis can be markedly enhanced by increased receptor activator of NF kappa-B ligand (RANKL) expression in PCa cells. Molecular mechanisms that account for the increased predilection of PCa for bone include increased bone turnover, promotion of PCa cell growth and survival in the bone environment, and recruitment of bystander dormant cells to participate in bone metastasis. The current study tests the hypothesis that PCa cells acquire high adhesion to bone matrix proteins, which controls PCa bone colonization, under the RANKL/RANK and AR axes. Methods: We used a highly bone metastatic RANKL-overexpressing LNCaP PCa cell line, LNCaPRANKL, as a model to pursue the molecular mechanisms underlying the increased adhesion of PCa cells to collagens. A three-dimensional (3-D) suspension PCa organoid model was developed. The functions of integrin α2 in cell adhesion and survival were evaluated by flow cytometry and western blot. AR expression and functionality were compared in 2-D monolayer versus 3-D suspension cultures using AR promoter- and PSA promoter-luciferase activity. AR role in cell adhesion was assessed using an adhesion assay. Results: LNCaPRANKL cells were shown to adhere tightly to ColI matrix through increased α2 integrin expression. This increased adhesion, concomitant with activation of the FAK and Akt pathways, was further enhanced by culturing LNCaPRANKL cells in 3-D suspension. Under the influence of 3-D suspension culture, AR was restored in LNCaPRANKL cells via downregulation of AP-4 transcription factor, and supported increased α2 integrin expression and adhesion to ColI. Conclusion: 3-D suspension culture and in vivo PCa tumor growth restore AR through downregulation of AP-4, enhancing integrin α2 expression and adhesion to ColI which is rich in bone matrices. The interactions of PCa with ColI, mediated by integrin α2 and AR expression, could be a key molecular event accounting for PCa bone metastasis. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
20. Expression dynamics of integrin α2, α3, and αV upon osteogenic differentiation of human mesenchymal stem cells
- Author
-
Min Kyu Kim, Jeeseung Kim, Kyoung Soo Kim, Se-Ri Seo, Chun Jeih Ryu, Young-Joo Jang, Hyosun Seo, and Hyun Min Lee
- Subjects
endocrine system ,Integrin ,Integrin alpha2 ,Medicine (miscellaneous) ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Flow cytometry ,Integrin αV ,lcsh:Biochemistry ,Human mesenchymal stem cells ,Downregulation and upregulation ,Osteogenesis ,Osteogenic differentiation ,medicine ,Humans ,lcsh:QD415-436 ,Cells, Cultured ,lcsh:R5-920 ,Osteoblasts ,medicine.diagnostic_test ,biology ,Chemistry ,Research ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Biology ,Integrin α3 ,Cell sorting ,Integrin α2 ,equipment and supplies ,Cell biology ,Adipogenesis ,Cancer cell ,biology.protein ,Molecular Medicine ,Monoclonal antibodies ,Stem cell ,lcsh:Medicine (General) - Abstract
Background The differentiation of human mesenchymal stem cells (hMSCs) into osteoblasts (OBs) is a prerequisite for bone formation. However, little is known about the definitive surface markers for OBs during osteogenesis. Methods To study the surface markers on OBs, we generated and used monoclonal antibodies (MAbs) against surface molecules on transforming growth factor-β1 (TGF-β1)-treated cancer cells. The generated MAbs were further selected toward expression changes on hMSCs cultured with TGF-β1/bone morphogenetic protein-2 (BMP-2) or osteogenic differentiation medium (ODM) by flow cytometry. Immunoprecipitation and mass spectrometry were performed to identify target antigens of selected MAbs. Expression changes of the target antigens were evaluated in hMSCs, human periodontal ligament cells (hPDLCs), and human dental pulp cells (hDPCs) during osteogenic and adipogenic differentiation by quantitative polymerase chain reaction (qPCR) and flow cytometry. hMSCs were also sorted by the MAbs using magnetic-activated cell sorting system, and osteogenic potential of sorted cells was evaluated via Alizarin Red S (ARS) staining and qPCR. Results The binding reactivity of MR14-E5, one of the MAbs, was downregulated in hMSCs with ODM while the binding reactivity of ER7-A7, ER7-A8, and MR1-B1 MAbs was upregulated. Mass spectrometry and overexpression identified that MR14-E5, ER7-A7/ER7-A8, and MR1-B1 recognized integrin α2, α3, and αV, respectively. Upon osteogenic differentiation of hMSCs, the expression of integrin α2 was drastically downregulated, but the expression of integrin α3 and αV was upregulated in accordance with upregulation of osteogenic markers. Expression of integrin α3 and αV was also upregulated in hPDLCs and hDPCs during osteogenic differentiation. Cell sorting showed that integrin αV-high hMSCs have a greater osteogenic potential than integrin αV-low hMSCs upon the osteogenic differentiation of hMSCs. Cell sorting further revealed that the surface expression of integrin αV is more dramatically induced even in integrin αV-low hMSCs. Conclusion These findings suggest that integrin α3 and αV induction is a good indicator of OB differentiation. These findings also shed insight into the expression dynamics of integrins upon osteogenic differentiation of hMSCs and provide the reason why different integrin ligands are required for OB differentiation of hMSCs.
- Published
- 2020
21. Overexpressed ITGA2 promotes malignant tumor aggression by up-regulating PD-L1 expression through the activation of the STAT3 signaling pathway
- Author
-
Ping Fan, Dan Li, Yan Sun, Dianyun Ren, Zibo Meng, Bo Wang, Heshui Wu, Jingyuan Zhao, Zhiqiang Liu, and Xin Jin
- Subjects
0301 basic medicine ,Male ,STAT3 Transcription Factor ,Cancer Research ,Angiogenesis ,medicine.medical_treatment ,Integrin alpha2 ,Biology ,medicine.disease_cause ,lcsh:RC254-282 ,Stat3 Signaling Pathway ,B7-H1 Antigen ,Metastasis ,Phosphorylation of STAT3 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Pancreatic cancer ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Programmed death-ligand 1 ,Phosphorylation ,Cancer ,Cell Proliferation ,Sequence Analysis, RNA ,Research ,Immunotherapy ,Hep G2 Cells ,Integrin α2 ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Pancreatic Neoplasms ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Carcinogenesis ,Neoplasm Transplantation ,Signal Transduction - Abstract
BackgroundRecent studies have reported that Integrin alpha 2 (ITGA2) plays an essential role in tumor cell proliferation, invasion, metastasis, and angiogenesis. An abnormally expressed ITGA2 correlates with unfavorable prognoses in multiple types of cancer. However, the specific mechanism of how ITGA2 contributes to tumorigenesis remains unclear.MethodsThe GEPIA web tool was used to find the clinical relevance of ITGA2 in cancer, and this significance was verified using Western blotting analysis of paired patient tissues and immunohistochemistry of the pancreatic cancer tissue. Functional assays, such as the MTS assay, colony formation assay, and transwell assay, were used to determine the biological role of ITGA2 in human cancer. The relationship between ITGA2 and programmed death-ligand 1 (PD-L1) was examined using Western blot analysis, RT-qPCR assay, and immunohistochemistry. The protein-protein interaction between ITGA2 and STAT3 was detected via co-immunoprecipitation.ResultsOur study showed that ITGA2 was markedly overexpressed in several malignant tumor cells and clinical tissues. Blocking ITGA2 inhibited the proliferation and invasion ability of cancer cells significantly, whereas overexpressed ITGA2 increased the degree of those processes considerably. Additionally, the RNA-seq assay indicated that ITGA2 transcriptionally regulated the expression of PD-L1 in pancreatic cancer. We also demonstrated that ITGA2 interacted with STAT3 and up-regulated the phosphorylation of STAT3; this interaction might involve the mechanism of ITGA2 inducing PD-L1 expression in cancer cells. Our results suggest that ITGA2 plays a critical role in cancer cell progression and the regulation of PD-L1 by activating the STAT3 pathway.ConclusionsWe identified a novel mechanism by which ITGA2 plays a critical role in modulating cancer immune response by transcriptionally increasing the expression of PD-L1 in cancer cells. Thus, targeting ITGA2 is an effective method to enhance the efficacy of checkpoint immunotherapy against cancer.
- Published
- 2019
22. Gene and Protein Expression Profiles in a Mouse Model of Collagen-Induced Arthritis
- Author
-
Ki Jong Rhee, Ho Joong Sung, and Sun Yeong Gwon
- Subjects
Male ,0301 basic medicine ,Proteome ,Microarray ,Integrin alpha2 ,Arthritis ,Biology ,collagen-induced arthritis ,integrin α2 ,Arthritis, Rheumatoid ,Mice ,03 medical and health sciences ,medicine ,Animals ,Humans ,Gene ,Oligonucleotide Array Sequence Analysis ,Autoimmune disease ,Gene Expression Profiling ,General Medicine ,medicine.disease ,proteome analysis ,Arthritis, Experimental ,Blot ,030104 developmental biology ,Mice, Inbred DBA ,Rheumatoid arthritis ,Disease Progression ,Cancer research ,biomarker ,Biomarker (medicine) ,Collagen ,microarray ,Biomarkers ,Research Paper - Abstract
The risk of rheumatoid arthritis (RA), an autoimmune disease, in the elderly population increases along with that of atherosclerosis, cardiovascular disease, type 2 diabetes, and Alzheimer's disease. Identifying specific biomarkers for RA can clarify the underlying molecular mechanisms and can aid diagnosis and patient care. To this end, the present study investigated the genes and proteins that are differentially expressed in RA using a mouse collagen-induced arthritis (CIA) model. We performed gene microarray and proteome array analyses using blood samples from the mice and found that 50 genes and 24 proteins were upregulated and 48 genes were downregulated by more than 2-fold in the CIA model relative to the control. The gene microarray and proteome array results were validated by evaluating the expression levels of select genes and proteins by real-time PCR and western blotting, respectively. We found that the level of integrin α2, which has not been previously reported as a biomarker of RA, was significantly increased in CIA mice as compared to controls. These findings provide a set of novel biomarkers that can be useful for diagnosing and evaluating the progression of RA.
- Published
- 2018
23. PRL-3 suppresses c-Fos and integrin α2 expression in ovarian cancer cells.
- Author
-
Hao Liu, Omer Al-aidaroos, Abdul Qader, Haihe Wang, Guo, Ke, Li, Jie, Hua Fei Zhang, and Zeng, Qi
- Subjects
- *
INTEGRINS , *OVARIAN cancer , *PHOSPHATASES , *CANCER cell migration , *IMMUNOHISTOCHEMISTRY , *CANCER invasiveness - Abstract
Background: Phosphatase of regenerating liver-3 (PRL-3), a protein tyrosine phosphatase, is highly expressed in multiple human cancers and strongly implicated in tumor progression and cancer metastasis. However, the mechanisms by which PRL-3 promotes cancer cell migration, invasion, and metastasis are not very well understood. In this study, we investigated the contribution and molecular mechanisms of PRL-3 in ovarian cancer progression. Methods: PRL-3 protein expression was detected on ovarian cancer tissue microarrays using immunohistochemistry. Stable PRL-3 depleted cell lines were generated using short hairpin RNA (shRNA) constructs. The migration and invasion potential of these cells were analyzed using Transwell and Matrigel assays, respectively. Immunoblotting and immunofluorescence were used to detect protein levels and distribution in PRL-3-ablated cells and the control cells. Cell morphology was observed with hematoxylin-eosin staining and transmission electron microscopy. Finally, PRL-3 -ablated and control cells were injected into nude mice for xenograft tumorigenicity assays. Results: Elevated PRL-3 expression was detected in 19% (26 out of 135) of human ovarian cancer patient samples, but not in normal ovary tissues (0 out of 14). Stable depletion of PRL-3 in A2780 ovarian cancer cells resulted in decreased migration ability and invasion activity compared with control parental A2780 cells. In addition, PRL-3 -ablated cells also exhibited flattened morphology and extended lamellipodia. To address the possible molecular basis for the altered phenotypes associated with PRL-3 down-regulation, we assessed the expression profiles of various proteins involved in cell-matrix adhesion. Depletion of PRL-3 dramatically enhanced both RNA and protein levels of the cell surface receptor integrin α2, but not its heterologous binding partner integrin β1. Inhibition of PRL-3 also correlated with elevated expression and phosphorylation of paxillin. A pronounced increase in the expression and activation of c-fos, a transcriptional activator of integrin α2, was observed in these PRL-3 knock-down cells. Moreover, forced expression of EGFP-PRL-3 resulted in the suppression of both integrin α2 and c-fos expression in A2780 cells. Significantly, using a xenograft tumor model, we observed a greatly reduced tumorigenicity of A2780 PRL-3 knock-down cells in vivo. Conclusions: These results suggest that PRL-3 plays a critical role in ovarian cancer tumorigenicity and maintaining the malignant phenotype. PRL-3 may inhibit c-fos transcriptional regulation of integrin α2 signaling. Our results strongly support a role for PRL-3 as a promising therapeutic target and potential early biomarker in ovarian cancer progression. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
24. Expression of Laminin ββ1 and Integrin αα2 in the Anterior Temporal Neocortex Tissue of Patients With Intractable Epilepsy.
- Author
-
Wu, Yuan, Wang, Xue-feng, Mo, Xue-an, Li, Jing-mei, Yuan, Jie, Zheng, Jin-ou, Feng, Yun, and Tang, Mei
- Subjects
- *
NEOCORTEX , *EPILEPSY , *IMMUNOHISTOCHEMISTRY , *IMMUNOFLUORESCENCE , *CELL membranes , *CYTOPLASM , *FLUORESCENCE - Abstract
We investigated the expression of laminin ββ1 and integrin αα2 in the anterior temporal neocortex tissue of patients with intractable epilepsy and explored the role of these molecules in the pathogenesis of this disease. Immunohistochemistry and immunofluorescence were used to test the expression of laminin ββ1 and integrin αα2 in samples (from the brain bank of our department, n == 32) of surgically removed anterior temporal neocortex tissues from intractable epilepsy patients, and the results were compared with those of controls ( n == 10). We found that laminin ββ1 and integrin αα2 protein expression was significantly increased in the anterior temporal neocortex as compared with controls (immunohistochemistry optical density: laminin ββ1 == 0.36 ±± 0.01 vs. 0.10 ±± 0.03 for control; integrin αα2 == 0.42 ±± 0.02 vs. 0.04 ±± 0.01 for control; p < .05). Immunofluorescence staining indicated that laminin ββ1 and integrin αα2 accumulated in the plasma membrane and cytoplasm, with strong fluorescence intensity in the anterior temporal neocortex tissue of patients with intractable epilepsy. Thus, our work demonstrates that laminin ββ1 and integrin αα2 expression is elevated in the anterior temporal neocortex tissue from patients with intractable epilepsy. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
25. Syndecan-2 overexpression regulates adhesion and migration through cooperation with integrin α2
- Author
-
Choi, Sojoong, Kim, Yeonhee, Park, Haein, Han, Inn-Oc, Chung, Eunkyung, Lee, Sung-Yul, Kim, Yong-Bae, Lee, Jung Weon, Oh, Eok-Soo, and Yi, Jae Youn
- Subjects
- *
PROTEOGLYCANS , *GENETIC regulation , *CELL adhesion , *CELL migration , *INTEGRINS , *CELLULAR control mechanisms , *CANCER invasiveness - Abstract
Abstract: Syndecan-2, a transmembrane heparan sulfate proteoglycan, is known to serve as an adhesion receptor, but details of the regulatory mechanism governing syndecan-2 cell adhesion and migration remain unclear. Here, we examined this regulatory mechanism, showing that overexpression of syndecan-2 enhanced collagen adhesion, cell migration and invasion of normal rat intestinal epithelial cells (RIE1), and increased integrin α2 expression levels. Interestingly, RIE1 cells transfected with either syndecan-2 or integrin α2 showed similar adhesion and migration patterns, and a function-blocking anti-integrin α2 antibody abolished syndecan-2-mediated adhesion and migration. Consistent with these findings, transfection of integrin α2 siRNA diminished syndecan-2-induced cell migration in HCT116 human colon cancer cells. Taken together, these results demonstrate a novel cooperation between syndecan-2 and integrin α2β1 in adhesion-mediated cell migration and invasion. This interactive dynamic might be a possible mechanism underlying the tumorigenic activities of colon cancer cells. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
26. Association of integrin α2 gene variants with ischemic stroke.
- Author
-
Matarin, Mar, Brown, W. Mark, Hardy, John A., Rich, Stephen S., Singleton, Andrew B., Brown Jr., Robert D., Brott, Thomas G., Worrall, Bradford B., and Meschia, James F.
- Subjects
- *
GENETIC polymorphisms , *ISCHEMIA , *GENES , *BLOOD flow , *CEREBRAL infarction , *GENETICS , *CEREBROVASCULAR disease - Abstract
Genetic variants in the gene encoding integrin α2 (ITGA2) have been reported to be associated with an increased risk for ischemic stroke. The purpose of this study was to investigate the association between haplotype-tagging single-nucleotide polymorphisms (tSNPs) in ITGA2 and risk of ischemic stroke in a collection of North American stroke cases and controls. The study included 484 cases and 263 controls. Thirteen tSNPs were genotyped. Association tests at and across each tSNP were performed, including haplotype association analysis. Secondary analyses considered stroke subtypes on the basis of Trial of Org 10172 in Acute Stroke Treatment (TOAST) criteria. We observed significant association between tSNP rs3756541 (additive model, odds ratio (OR), 1.49; 95% confidence interval (CI), 1.11 to 2.04; P=0.009) and disease and a trend toward association at rs2303124 (recessive model, OR, 1.56; 95% CI, 1.05 to 2.33; P=0.03). These associations remained significant in the haplotype analyses. The associated tSNPs did not distinguish stroke etiology after application of TOAST criteria. Our results suggest that genetic variability within ITGA2 may confer risk for ischemic stroke independent of conventional risk factors. These results provide additional support for a role for platelet receptor genes in the pathogenesis of ischemic stroke of diverse subtypes.Journal of Cerebral Blood Flow & Metabolism (2008) 28, 81–89; doi:10.1038/sj.jcbfm.9600508; published online 30 May 2007 [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
27. Targeting platelet GPVI plus rt-PA administration but not α2β1-mediated collagen binding protects against ischemic brain damage in mice
- Author
-
Michael K, Schuhmann, Peter, Kraft, Michael, Bieber, Alexander M, Kollikowski, Harald, Schulze, Bernhard, Nieswandt, Mirko, Pham, David, Stegner, and Guido, Stoll
- Subjects
Male ,Platelet Membrane Glycoproteins ,Protective Agents ,Antibodies ,glycoprotein VI ,lcsh:Chemistry ,Mice ,ischemic stroke ,Animals ,ddc:610 ,transient middle cerebral artery occlusion ,lcsh:QH301-705.5 ,integrin α2 ,Communication ,intracranial bleeding ,Brain ,Infarction, Middle Cerebral Artery ,Recombinant Proteins ,Mice, Inbred C57BL ,Stroke ,recombinant tissue-type plasminogen activator ,lcsh:Biology (General) ,lcsh:QD1-999 ,Tissue Plasminogen Activator ,Collagen ,Integrin alpha2beta1 - Abstract
Platelet collagen interactions at sites of vascular injuries predominantly involve glycoprotein VI (GPVI) and the integrin α2β1. Both proteins are primarily expressed on platelets and megakaryocytes whereas GPVI expression is also shown on endothelial and integrin α2β1 expression on epithelial cells. We recently showed that depletion of GPVI improves stroke outcome without increasing the risk of cerebral hemorrhage. Genetic variants associated with higher platelet surface integrin α2 (ITGA2) receptor levels have frequently been found to correlate with an increased risk of ischemic stroke in patients. However until now, no preclinical stroke study has addressed whether platelet integrin α2β1 contributes to the pathophysiology of ischemia/reperfusion (I/R) injury. Focal cerebral ischemia was induced in C57BL/6 and Itga2−/− mice by a 60 min transient middle cerebral artery occlusion (tMCAO). Additionally, wild-type animals were pretreated with anti-GPVI antibody (JAQ1) or Fab fragments of a function blocking antibody against integrin α2β1 (LEN/B). In anti-GPVI treated animals, intravenous (IV) recombinant tissue plasminogen activator (rt-PA) treatment was applied immediately prior to reperfusion. Stroke outcome, including infarct size and neurological scoring was determined on day 1 after tMCAO. We demonstrate that targeting the integrin α2β1 (pharmacologic; genetic) did neither reduce stroke size nor improve functional outcome on day 1 after tMCAO. In contrast, depletion of platelet GPVI prior to stroke was safe and effective, even when combined with rt-PA treatment. Our results underscore that GPVI, but not ITGA2, is a promising and safe target in the setting of ischemic stroke.
- Published
- 2019
28. Integrin α2 marks a niche of trophoblast progenitor cells in first trimester human placenta
- Author
-
Lee, Cheryl QE, Turco, Margherita Y, Gardner, Lucy, Simons, Benjamin D, Hemberger, Myriam, Moffett, Ashley, Lee, Cheryl QE [0000-0003-3542-0833], Turco, Margherita Y [0000-0002-3380-7375], Gardner, Lucy [0000-0003-1735-2005], Simons, Benjamin D [0000-0002-3875-7071], Hemberger, Myriam [0000-0003-3332-6958], Moffett, Ashley [0000-0002-8388-9073], and Apollo - University of Cambridge Repository
- Subjects
Placenta ,Stem Cells ,Integrin alpha2 ,Integrin α2 ,Lineage tracing ,Placentation ,Trophoblasts ,Pregnancy Trimester, First ,Pregnancy ,embryonic structures ,Humans ,Female ,Receptor, Notch1 ,reproductive and urinary physiology ,Biomarkers ,Progenitor ,Cell Proliferation ,Signal Transduction - Abstract
During pregnancy the trophoblast cells of the placenta are the only fetal cells in direct contact with maternal blood and decidua. Their functions include transport of nutrients and oxygen, secretion of pregnancy hormones, remodelling of the uterine arteries, and communicating with maternal cells. Despite the importance of trophoblast cells in placental development and successful pregnancy, little is known about the identity, location and differentiation of human trophoblast progenitors. We identify a proliferative trophoblast niche at the base of the cytotrophoblast cell columns in first trimester placentas that is characterised by integrin α2 (ITGA2) expression. Pulse-chase experiments with 5-iodo-2'-deoxyuridine indicate that these cells might contribute to both villous (VCT) and extravillous (EVT) lineages. These proliferating trophoblast cells can be isolated by flow cytometry using ITGA2 as a marker and express genes from both VCT and EVT. Microarray expression analysis shows that ITAG2+ cells display a unique transcriptional signature, including genes involved in NOTCH signalling, and exhibit a combination of epithelial and mesenchymal characteristics. ITGA2 thus marks a niche allowing the study of pure populations of trophoblast progenitor cells.
- Published
- 2018
29. Collagen Assembly at the Cell Surface: Dogmas Revisited.
- Author
-
Musiime, Moses, Chang, Joan, Hansen, Uwe, Kadler, Karl E., Zeltz, Cédric, Gullberg, Donald, and Rivero, Francisco
- Subjects
- *
COLLAGEN , *EXTRACELLULAR matrix , *DOGMA , *CELL culture , *TISSUE wounds , *INTEGRINS , *FIBRONECTINS , *WOUND healing - Abstract
With the increased awareness about the importance of the composition, organization, and stiffness of the extracellular matrix (ECM) for tissue homeostasis, there is a renewed need to understand the details of how cells recognize, assemble and remodel the ECM during dynamic tissue reorganization events. Fibronectin (FN) and fibrillar collagens are major proteins in the ECM of interstitial matrices. Whereas FN is abundant in cell culture studies, it is often only transiently expressed in the acute phase of wound healing and tissue regeneration, by contrast fibrillar collagens form a persistent robust scaffold in healing and regenerating tissues. Historically fibrillar collagens in interstitial matrices were seen merely as structural building blocks. Cell anchorage to the collagen matrix was thought to be indirect and occurring via proteins like FN and cell surface-mediated collagen fibrillogenesis was believed to require a FN matrix. The isolation of four collagen-binding integrins have challenged this dogma, and we now know that cells anchor directly to monomeric forms of fibrillar collagens via the α1β1, α2β1, α10β1 and α11β1 integrins. The binding of these integrins to the mature fibrous collagen matrices is more controversial and depends on availability of integrin-binding sites. With increased awareness about the importance of characterizing the total integrin repertoire on cells, including the integrin collagen receptors, the idea of an absolute dependence on FN for cell-mediated collagen fibrillogenesis needs to be re-evaluated. We will summarize data suggesting that collagen-binding integrins in vitro and in vivo are perfectly well suited for nucleating and supporting collagen fibrillogenesis, independent of FN. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
30. Hierarchical Micro-Nano Topography Promotes Cell Adhesion and Osteogenic Differentiation via Integrin α2-PI3K-AKT Signaling Axis.
- Author
-
Zheng H, Tian Y, Gao Q, Yu Y, Xia X, Feng Z, Dong F, Wu X, and Sui L
- Abstract
Surface topography dictates important aspects of cell biological behaviors. In our study, hierarchical micro-nano topography (SLM-AHT) with micro-scale grooves and nano-scale pores was fabricated and compared with smooth topography (S) and irregular micro-scale topography (SLA) surfaces to investigate mechanism involved in cell-surface interactions. Integrin α2 had a higher expression level on SLM-AHT surface compared with S and SLA surfaces, and the expression levels of osteogenic markers icluding Runx2, Col1a1, and Ocn were concomitantly upregulated on SLM-AHT surface. Moreover, formation of mature focal adhesions were significantly enhanced in SLM-AHT group. Noticablely, silencing integrin α2 could wipe out the difference of osteogenic gene expression among surfaces with different topography, indicating a crucial role of integrin α2 in topography induced osteogenic differentiation. In addition, PI3K-AKT signaling was proved to be regulated by integrin α2 and consequently participate in this process. Taken together, our findings illustrated that integrin α2-PI3K-AKT signaling axis plays a key role in hierarchical micro-nano topography promoting cell adhesion and osteogenic differentiation., (Copyright © 2020 Zheng, Tian, Gao, Yu, Xia, Feng, Dong, Wu and Sui.)
- Published
- 2020
- Full Text
- View/download PDF
31. Targeting Platelet GPVI Plus rt-PA Administration but Not α2β1-Mediated Collagen Binding Protects against Ischemic Brain Damage in Mice.
- Author
-
Schuhmann, Michael K., Kraft, Peter, Bieber, Michael, Kollikowski, Alexander M., Schulze, Harald, Nieswandt, Bernhard, Pham, Mirko, Stegner, David, and Stoll, Guido
- Subjects
- *
HEMORRHAGE , *COLLAGEN , *PATHOLOGICAL physiology , *PLASMINOGEN , *REPERFUSION - Abstract
Platelet collagen interactions at sites of vascular injuries predominantly involve glycoprotein VI (GPVI) and the integrin α2β1. Both proteins are primarily expressed on platelets and megakaryocytes whereas GPVI expression is also shown on endothelial and integrin α2β1 expression on epithelial cells. We recently showed that depletion of GPVI improves stroke outcome without increasing the risk of cerebral hemorrhage. Genetic variants associated with higher platelet surface integrin α2 (ITGA2) receptor levels have frequently been found to correlate with an increased risk of ischemic stroke in patients. However until now, no preclinical stroke study has addressed whether platelet integrin α2β1 contributes to the pathophysiology of ischemia/reperfusion (I/R) injury. Focal cerebral ischemia was induced in C57BL/6 and Itga2−/− mice by a 60 min transient middle cerebral artery occlusion (tMCAO). Additionally, wild-type animals were pretreated with anti-GPVI antibody (JAQ1) or Fab fragments of a function blocking antibody against integrin α2β1 (LEN/B). In anti-GPVI treated animals, intravenous (IV) recombinant tissue plasminogen activator (rt-PA) treatment was applied immediately prior to reperfusion. Stroke outcome, including infarct size and neurological scoring was determined on day 1 after tMCAO. We demonstrate that targeting the integrin α2β1 (pharmacologic; genetic) did neither reduce stroke size nor improve functional outcome on day 1 after tMCAO. In contrast, depletion of platelet GPVI prior to stroke was safe and effective, even when combined with rt-PA treatment. Our results underscore that GPVI, but not ITGA2, is a promising and safe target in the setting of ischemic stroke. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
32. Novel method for the isolation and characterisation of the putative prostatic stem cell
- Author
-
Michael D Brown, Noel W. Clarke, Vijay A C Ramani, Claire A Hart, N. J. R. George, Rupesh Bhatt, and Paul E Gilmore
- Subjects
Male ,PCA3 ,Hoechst ,medicine.medical_treatment ,Cell Culture Techniques ,Biophysics ,Stem cells ,Biology ,Stem cell marker ,Resting Phase, Cell Cycle ,Pathology and Forensic Medicine ,Prostate cancer ,Endocrinology ,Side population ,Prostate ,Cancer stem cell ,medicine ,Humans ,Fluorescent Dyes ,Transurethral resection of the prostate ,Stem Cells ,Cell Cycle ,G1 Phase ,Epithelial Cells ,Cell Biology ,Hematology ,Integrin α2 ,Calcium Channel Blockers ,Flow Cytometry ,Hematopoietic Stem Cells ,medicine.disease ,Phenotype ,medicine.anatomical_structure ,Verapamil ,Immunology ,Cancer research ,Keratins ,Leukocyte Common Antigens ,Benzimidazoles ,Prostate epithelial cells ,Stem cell ,Propidium - Abstract
Background Prostate stem cells, responsible for the development, maturation, and function of the prostate, have been implicated in the aetiology of both benign prostate hyperplasia (BPH) and prostate cancer (CaP). However, research has been hampered by the lack of a definitive stem cell marker. We have adapted the protocol for differential Hoechst 33342 uptake by hemopoietic stem cells to enable isolation of putative stem cells from the prostate. Methods Prostate epithelial cells isolated from prostate tissue obtained from patients with BPH after transurethral resection of the prostate were stained with Hoechst 33342. The Hoechst 33342 Red/Blue flow cytometry profile was then determined. Hoechst 33342 and Pyronin Y staining was used to determined the cell cycle status. Results A verapamil-sensitive side population (SP) can be isolated from primary prostate tissue accounting for 1.38% ± 0.07% of prostate epithelial cells. Cell cycle analysis of this SP population revealed that the majority of SP cells are in either G0 (12.38 ± 0.31%) or G1 (63.19 ± 2.13%). Conclusions The Hoechst 33342 dye efflux protocol can be adapted for the isolation of a SP from primary prostate tissue. Cytometry Part A 54A:89–99, 2003. © 2003 Wiley-Liss, Inc.
- Published
- 2003
33. Polymorphism in Integrin ITGA2 is Associated with Ischemic Stroke and Altered Serum Cholesterol in Chinese Individuals
- Author
-
Shao lan Zhang, Jian Xia Lu, Zhong Qian Lu, Wan Xiang Wang, Zheng Ping Chen, and Juan Zhi
- Subjects
medicine.medical_specialty ,lcsh:Medicine ,Integrin β3 ,Health Care Sciences and Services ,Internal medicine ,Genotype ,ischemic stroke ,medicine ,Allele ,Sağlık Bilimleri ve Hizmetleri ,Allele frequency ,business.industry ,lcsh:R ,Gene polymorphism ,Case-control study ,Gene polymorphism,Integrin α2,Integrin β3,ischemic stroke,plasma lipid ,General Medicine ,Odds ratio ,Integrin α2 ,plasma lipid ,Endocrinology ,Relative risk ,Immunology ,Original Article ,business ,Lipoprotein - Abstract
Background: Recent studies have reported contrasting results regarding the association of polymorphisms in two integrin genes, ITGA2 and ITGB3, with ischemic stroke. Aims: The present study aimed to investigate the correlation between the ITGA2 C807T and ITGB3 T176C polymorphic loci with ischemic stroke, as well as plasma lipid and lipoprotein levels. Study Design: Case control study. Methods: Human venous blood samples were collected from patients admitted for ischemic stroke (n=350, ‘patients') and healthy individuals (n=300, ‘controls'). Blood was genotyped at these loci by polymerase chain reaction-restriction fragment length polymorphism. Plasma lipid and lipoprotein levels were measured by routine enzymatic, masking, and turbidimetry methods. Results: As expected, total cholesterol, triglycerides, and low-density lipoprotein were all significantly higher in patients than in controls (p
- Published
- 2014
34. Genetic Polymorphisms of Platelet Adhesive Molecules: Association with Breast Cancer Risk and Clinical Presentation
- Author
-
Ayala, Francisco, Corral, Javier, González-Conejero, Rocío, Sánchez, Ignacio, María Moraleda, José, and Vicente, Vicente
- Published
- 2003
- Full Text
- View/download PDF
35. Regulation and functions of integrin α2 in cell adhesion and disease.
- Author
-
Adorno-Cruz V and Liu H
- Abstract
Integrins are cell adhesion molecules that are composed of an alpha (α) subunit and a beta (β) subunit with affinity for different extracellular membrane components. The integrin family includes 24 known members that actively regulate cellular growth, differentiation, and apoptosis. Each integrin heterodimer has a particular function in defined contexts as well as some partially overlapping features with other members in the family. As many reviews have covered the general integrin family in molecular and cellular studies in life science, this review will focus on the specific regulation, function, and signaling of integrin α2 subunit (CD49b, VLA-2; encoded by the gene ITGA2 ) in partnership with β1 (CD29) subunit in normal and cancer cells. Its roles in cell adhesion, cell motility, angiogenesis, stemness, and immune/blood cell regulations are discussed. The pivotal role of integrin α2 in many diseases such as cancer suggests its potential to be used as a novel therapeutic target.
- Published
- 2018
- Full Text
- View/download PDF
36. Cantharidin and norcantharidin inhibit the ability of MCF-7 cells to adhere to platelets via protein kinase C pathway-dependent downregulation of α2 integrin
- Author
-
Kai Chen, Zhenyu Li, Xin Xie, Min Tao, Qiong-Yan Zhang, Fei-Ran Gong, Wei Li, Yixiang Mao, Liu-Mei Shou, Lian Lian, and Kesheng Dai
- Subjects
Blood Platelets ,Cancer Research ,Blotting, Western ,Integrin alpha2 ,Down-Regulation ,Apoptosis ,Breast Neoplasms ,Biology ,cantharidin and norcantharidin ,Real-Time Polymerase Chain Reaction ,chemistry.chemical_compound ,Platelet Adhesiveness ,breast cancer ,Cell Adhesion ,Humans ,RNA, Messenger ,Enzyme Inhibitors ,RNA, Small Interfering ,Cell adhesion ,Protein Kinase C ,Cell Proliferation ,platelet ,Cantharidin ,Wound Healing ,Norcantharidin ,integrin α2 ,Cell growth ,Reverse Transcriptase Polymerase Chain Reaction ,protein phosphatase 2A ,Transcription Factor RelA ,General Medicine ,Articles ,Cell cycle ,Bridged Bicyclo Compounds, Heterocyclic ,Flow Cytometry ,Surface coating ,Oncology ,MCF-7 ,chemistry ,Cancer cell ,Cancer research ,MCF-7 Cells ,Female - Abstract
Cancer metastasis is a highly coordinated and dynamic multistep process in which cancer cells interact with a variety of host cells. Morphological studies have documented the association of circulating tumor cells with host platelets, where a surface coating of platelets protects tumor cells from mechanical trauma and the immune system. Cantharidin is an active constituent of mylabris, a traditional Chinese medicine. Cantharidin and norcantharidin are potent protein phosphatase 2A (PP2A) inhibitors that exhibit in vitro and in vivo antitumor activity against several types of cancer, including breast cancer. We investigated whether cantharidin and norcantharidin could repress the ability of MCF-7 breast cancer cells to adhere to platelets. Using MTT, clone formation, apoptosis, adhesion and wound-healing assays, we found that cantharidin and norcantharidin induced apoptosis and repressed MCF-7 cell growth, adhesion and migration. Moreover, we developed a flow cytometry-based analysis of tumor cell adhesion to platelets. We proved that cantharidin and norcantharidin repressed MCF-7 cell adhesion to platelets through downregulation of α2 integrin, an adhesion molecule present on the surface of cancer cells. The repression of α2 integrin expression was found to be executed through the protein kinase C pathway, the activation of which could have been due to PP2A inhibition.
- Published
- 2013
37. Influence of collagen-based integrin α 1 and α 2 mediated signaling on human mesenchymal stem cell osteogenesis in three dimensional contexts.
- Author
-
Becerra-Bayona SM, Guiza-Arguello VR, Russell B, Höök M, and Hahn MS
- Subjects
- Biomarkers metabolism, Humans, Hydrogels pharmacology, MAP Kinase Signaling System drug effects, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells drug effects, Osteopontin metabolism, Polyethylene Glycols pharmacology, Protein Subunits metabolism, Sp7 Transcription Factor metabolism, Tensile Strength, Collagen metabolism, Integrin alpha1 metabolism, Integrin alpha2 metabolism, Mesenchymal Stem Cells metabolism, Osteogenesis drug effects, Signal Transduction
- Abstract
Collagen I interactions with integrins α
1 and α2 are known to support human mesenchymal stem cell (hMSC) osteogenesis. Nonetheless, elucidating the relative impact of specific integrin interactions has proven challenging, in part due to the complexity of native collagen. In the present work, we employed two collagen-mimetic proteins-Scl2-2 and Scl2-3- to compare the osteogenic effects of integrin α1 versus α2 signaling. Scl2-2 and Scl2-3 were both derived from Scl2-1, a triple helical protein lacking known cell adhesion, cytokine binding, and matrix metalloproteinase sites. However, Scl2-2 and Scl2-3 were each engineered to display distinct collagen-based cell adhesion motifs: GFPGER (binding integrins α1 and α2 ) or GFPGEN (binding only integrin α1 ), respectively. hMSCs were cultured within poly(ethylene glycol) (PEG) hydrogels containing either Scl2-2 or Scl2-3 for 2 weeks. PEG-Scl2-2 gels were associated with increased hMSC osterix expression, osteopontin production, and calcium deposition relative to PEG-Scl2-3 gels. These data indicate that integrin α2 signaling may have an increased osteogenic effect relative to integrin α1 . Since p38 is activated by integrin α2 but not by integrin α1 , hMSCs were further cultured in PEG-Scl2-2 hydrogels in the presence of a p38 inhibitor. Results suggest that p38 activity may play a key role in collagen-supported hMSC osteogenesis. This knowledge can be used toward the rational design of scaffolds which intrinsically promote hMSC osteogenesis. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 2594-2604, 2018., (Copyright © 2018 Wiley Periodicals, Inc.)- Published
- 2018
- Full Text
- View/download PDF
38. Polymorphism in Integrin ITGA2 is Associated with Ischemic Stroke and Altered Serum Cholesterol in Chinese Individuals.
- Author
-
Lu JX, Lu ZQ, Zhang SL, Zhi J, Chen ZP, and Wang WX
- Abstract
Background: Recent studies have reported contrasting results regarding the association of polymorphisms in two integrin genes, ITGA2 and ITGB3, with ischemic stroke., Aims: The present study aimed to investigate the correlation between the ITGA2 C807T and ITGB3 T176C polymorphic loci with ischemic stroke, as well as plasma lipid and lipoprotein levels., Study Design: Case control study., Methods: Human venous blood samples were collected from patients admitted for ischemic stroke (n=350, 'patients') and healthy individuals (n=300, 'controls'). Blood was genotyped at these loci by polymerase chain reaction-restriction fragment length polymorphism. Plasma lipid and lipoprotein levels were measured by routine enzymatic, masking, and turbidimetry methods., Results: As expected, total cholesterol, triglycerides, and low-density lipoprotein were all significantly higher in patients than in controls (p<0.05). Genotype and allele frequencies of ITGA2 C807T were significantly different between patients and controls (p<0.05), but no difference was detected in genotype or allele frequencies for ITGA3 T176C. For ITGA-2, the T allele conferred a 1.226 times higher relative risk of ischemic stroke than the C allele (odds ratio=1.226, 95% confidence interval=1.053-1.428). Similarly, total cholesterol was higher in T allele carriers than in non-carriers (p<0.05)., Conclusion: ITGA2 C807T polymorphism is associated with ischemic stroke, with the T allele acting as a susceptibility allele that appears to confer increased cholesterol levels.
- Published
- 2014
- Full Text
- View/download PDF
39. PRL-3 suppresses c-Fos and integrin α2 expression in ovarian cancer cells
- Author
-
Qi Zeng, Ke Guo, Hua Fei Zhang, Haihe Wang, Abdul Qader Omer Al-aidaroos, Jie Li, and Hao Liu
- Subjects
endocrine system ,Cancer Research ,Adhesion molecule ,endocrine system diseases ,Transplantation, Heterologous ,Integrin alpha2 ,Biology ,Cell morphology ,Metastasis ,Small hairpin RNA ,PRL-3 phosphatase ,Mice ,Cell Movement ,Cell Line, Tumor ,medicine ,Genetics ,Animals ,Humans ,Cell migration ,Neoplasm Invasiveness ,c-fos transcription factor ,Ovarian Neoplasms ,Matrigel ,Cancer metastasis ,Integrin α2 ,medicine.disease ,Neoplasm Proteins ,Tumor Burden ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,Cell Transformation, Neoplastic ,Oncology ,Tumor progression ,Gene Knockdown Techniques ,Cancer research ,Female ,Paxillin ,Protein Tyrosine Phosphatases ,Ovarian cancer ,Proto-Oncogene Proteins c-fos ,hormones, hormone substitutes, and hormone antagonists ,Research Article - Abstract
Background Phosphatase of regenerating liver-3 (PRL-3), a protein tyrosine phosphatase, is highly expressed in multiple human cancers and strongly implicated in tumor progression and cancer metastasis. However, the mechanisms by which PRL-3 promotes cancer cell migration, invasion, and metastasis are not very well understood. In this study, we investigated the contribution and molecular mechanisms of PRL-3 in ovarian cancer progression. Methods PRL-3 protein expression was detected on ovarian cancer tissue microarrays using immunohistochemistry. Stable PRL-3 depleted cell lines were generated using short hairpin RNA (shRNA) constructs. The migration and invasion potential of these cells were analyzed using Transwell and Matrigel assays, respectively. Immunoblotting and immunofluorescence were used to detect protein levels and distribution in PRL-3-ablated cells and the control cells. Cell morphology was observed with hematoxylin-eosin staining and transmission electron microscopy. Finally, PRL-3-ablated and control cells were injected into nude mice for xenograft tumorigenicity assays. Results Elevated PRL-3 expression was detected in 19% (26 out of 135) of human ovarian cancer patient samples, but not in normal ovary tissues (0 out of 14). Stable depletion of PRL-3 in A2780 ovarian cancer cells resulted in decreased migration ability and invasion activity compared with control parental A2780 cells. In addition, PRL-3-ablated cells also exhibited flattened morphology and extended lamellipodia. To address the possible molecular basis for the altered phenotypes associated with PRL-3 down-regulation, we assessed the expression profiles of various proteins involved in cell-matrix adhesion. Depletion of PRL-3 dramatically enhanced both RNA and protein levels of the cell surface receptor integrin α2, but not its heterologous binding partner integrin β1. Inhibition of PRL-3 also correlated with elevated expression and phosphorylation of paxillin. A pronounced increase in the expression and activation of c-fos, a transcriptional activator of integrin α2, was observed in these PRL-3 knock-down cells. Moreover, forced expression of EGFP-PRL-3 resulted in the suppression of both integrin α2 and c-fos expression in A2780 cells. Significantly, using a xenograft tumor model, we observed a greatly reduced tumorigenicity of A2780 PRL-3 knock-down cells in vivo. Conclusions These results suggest that PRL-3 plays a critical role in ovarian cancer tumorigenicity and maintaining the malignant phenotype. PRL-3 may inhibit c-fos transcriptional regulation of integrin α2 signaling. Our results strongly support a role for PRL-3 as a promising therapeutic target and potential early biomarker in ovarian cancer progression.
- Full Text
- View/download PDF
40. Induction of integrin α2 in a highly bone metastatic human prostate cancer cell line: roles of RANKL and AR under three-dimensional suspension culture
- Author
-
Shabnam Ziaee and Leland W.K. Chung
- Subjects
Male ,medicine.medical_specialty ,Cancer Research ,Cell Survival ,Integrin ,Cell Culture Techniques ,Integrin alpha2 ,Bone Neoplasms ,Biology ,urologic and male genital diseases ,Collagen Type I ,Bone remodeling ,Mice ,Internal medicine ,3-D culture ,Cell Line, Tumor ,LNCaP ,medicine ,Cell Adhesion ,Animals ,Humans ,Cell adhesion ,Prostate cancer ,Cell growth ,Research ,RANK Ligand ,Bone metastasis ,Prostatic Neoplasms ,Integrin α2 ,medicine.disease ,3. Good health ,AP-4 ,Gene Expression Regulation, Neoplastic ,Endocrinology ,Oncology ,RANKL ,Cell culture ,Receptors, Androgen ,Androgen Receptor ,Cancer research ,biology.protein ,Molecular Medicine ,Collagen ,Signal Transduction - Abstract
Background Prostate cancer (PCa) bone metastasis can be markedly enhanced by increased receptor activator of NF kappa-B ligand (RANKL) expression in PCa cells. Molecular mechanisms that account for the increased predilection of PCa for bone include increased bone turnover, promotion of PCa cell growth and survival in the bone environment, and recruitment of bystander dormant cells to participate in bone metastasis. The current study tests the hypothesis that PCa cells acquire high adhesion to bone matrix proteins, which controls PCa bone colonization, under the RANKL/RANK and AR axes. Methods We used a highly bone metastatic RANKL-overexpressing LNCaP PCa cell line, LNCaPRANKL, as a model to pursue the molecular mechanisms underlying the increased adhesion of PCa cells to collagens. A three-dimensional (3-D) suspension PCa organoid model was developed. The functions of integrin α2 in cell adhesion and survival were evaluated by flow cytometry and western blot. AR expression and functionality were compared in 2-D monolayer versus 3-D suspension cultures using AR promoter- and PSA promoter-luciferase activity. AR role in cell adhesion was assessed using an adhesion assay. Results LNCaPRANKL cells were shown to adhere tightly to ColI matrix through increased α2 integrin expression. This increased adhesion, concomitant with activation of the FAK and Akt pathways, was further enhanced by culturing LNCaPRANKL cells in 3-D suspension. Under the influence of 3-D suspension culture, AR was restored in LNCaPRANKL cells via downregulation of AP-4 transcription factor, and supported increased α2 integrin expression and adhesion to ColI. Conclusion 3-D suspension culture and in vivo PCa tumor growth restore AR through downregulation of AP-4, enhancing integrin α2 expression and adhesion to ColI which is rich in bone matrices. The interactions of PCa with ColI, mediated by integrin α2 and AR expression, could be a key molecular event accounting for PCa bone metastasis. Electronic supplementary material The online version of this article (doi:10.1186/1476-4598-13-208) contains supplementary material, which is available to authorized users.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.