1
|
Gillani SMH, Mughal A, Khan RAA, Nawaz MH, Razzaq Z, Ismat MS, Hussain R, Wadood A, Ahmed S, Minhas B, Abbas M, Vayalpurayil T, Rehman MAU. Development of hybrid polyvinylpyrrolidone/carboxymethyl cellulose/collagen incorporated oregano scaffolds via direct ink write printing for potential wound healing applications. Int J Biol Macromol 2024; 278:134528. [PMID: 39111499 DOI: 10.1016/j.ijbiomac.2024.134528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/08/2024] [Accepted: 08/04/2024] [Indexed: 08/20/2024]
Abstract
Additive manufacturing can develop regenerative scaffolds for wound healing. 3D printing offers meticulous porosity, mechanical integrity, cell adhesion and cost-effectiveness. Herein, we prepared ink composed of carboxymethyl cellulose (CMC), polyvinylpyrrolidone (PVP), collagen, and oregano extract for the fabrication of tissue constructs. The blend was optimized to form a homogeneous ink and rheological characterization demonstrated shear thinning behavior. The scaffolds were printed using Direct Ink Write (DIW) at a flow speed of 4 mm3/s and a layer height of 0.18 mm. The fabricated scaffolds demonstrated an ultimate tensile strength (UTS) and toughness of 730 KPa and 2.72 MJ/m3, respectively. Scanning Electron Microscopy (SEM) revealed an average pore size of 300 ± 30 μm. Fourier transform infrared spectroscopy (FTIR) analysis confirmed that all materials were present. The contact angle of the composite scaffold was 68° ± 1°. Moreover, the scaffolds presented 82 % mass loss (degradation) in phosphate buffer saline (PBS) over 14 days. The composite scaffold exhibited inhibition zones of 9 mm and 12 mm against Staphylococcus aureus and Escherichia coli, respectively. The PVP/CMC/collagen/oregano 3D printed scaffolds exhibited excellent biocompatibility with the mesenchymal stem cells and humman dermal fibroblast cells, confirmed by water-soluble tetrazolium - 8 (WST-8) assay (test conducted for 7 days). The enhanced angiogenic potential of said scaffold was assesed by release of vascular endothelial growth factor followed by further validation through in-vivo CAM assay. Thus, confirming suitability for the potential wound healing application.
Collapse
Affiliation(s)
- Syed Muneeb Haider Gillani
- Center of Excellence in Biomaterials and Tissue Engineering, Materials Science and Engineering Department Government Collage University, 54000 Lahore, Pakistan
| | - Awab Mughal
- Center of Excellence in Biomaterials and Tissue Engineering, Materials Science and Engineering Department Government Collage University, 54000 Lahore, Pakistan
| | - Raja Aqib Akmal Khan
- Department of Materials Science & Engineering, Institute of Space Technology, 44000 Islamabad, Pakistan
| | - Muhammad Haseeb Nawaz
- Department of Materials Science & Engineering, Institute of Space Technology, 44000 Islamabad, Pakistan
| | - Zohaib Razzaq
- Department of Materials Science & Engineering, Institute of Space Technology, 44000 Islamabad, Pakistan
| | - Muhammad Sameet Ismat
- Center of Excellence in Biomaterials and Tissue Engineering, Materials Science and Engineering Department Government Collage University, 54000 Lahore, Pakistan
| | - Rabia Hussain
- Center of Excellence in Biomaterials and Tissue Engineering, Materials Science and Engineering Department Government Collage University, 54000 Lahore, Pakistan
| | - Abdul Wadood
- Department of Materials Science & Engineering, Institute of Space Technology, 44000 Islamabad, Pakistan
| | - Sheraz Ahmed
- Department of Materials Science & Engineering, Institute of Space Technology, 44000 Islamabad, Pakistan
| | - Badar Minhas
- Center of Excellence in Biomaterials and Tissue Engineering, Materials Science and Engineering Department Government Collage University, 54000 Lahore, Pakistan.
| | - Mohamed Abbas
- Central Labs, King Khalid University, AlQura'a, Abha, P.O. Box 960, Saudi Arabia; Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
| | - Thafasalijyas Vayalpurayil
- Central Labs, King Khalid University, AlQura'a, Abha, P.O. Box 960, Saudi Arabia; Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
| | - Muhammad Atiq Ur Rehman
- Center of Excellence in Biomaterials and Tissue Engineering, Materials Science and Engineering Department Government Collage University, 54000 Lahore, Pakistan; Department of Materials Science & Engineering, Institute of Space Technology, 44000 Islamabad, Pakistan.
| |
Collapse
|
2
|
Harada M, Matsuu A, Park ES, Inoue Y, Uda A, Kaku Y, Okutani A, Posadas-Herrera G, Ishijima K, Inoue S, Maeda K. Construction of Vero cell-adapted rabies vaccine strain by five amino acid substitutions in HEP-Flury strain. Sci Rep 2024; 14:12559. [PMID: 38822013 PMCID: PMC11143356 DOI: 10.1038/s41598-024-63337-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024] Open
Abstract
Rabies virus (RABV) causes fatal neurological disease. Pre-exposure prophylaxis (PrEP) and post-exposure prophylaxis (PEP) using inactivated-virus vaccines are the most effective measures to prevent rabies. In Japan, HEP-Flury, the viral strain, used as a human rabies vaccine, has historically been propagated in primary fibroblast cells derived from chicken embryos. In the present study, to reduce the cost and labor of vaccine production, we sought to adapt the original HEP-Flury (HEP) to Vero cells. HEP was repeatedly passaged in Vero cells to generate ten- (HEP-10V) and thirty-passaged (HEP-30V) strains. Both HEP-10V and HEP-30V grew significantly better than HEP in Vero cells, with virulence and antigenicity similar to HEP. Comparison of the complete genomes with HEP revealed three non-synonymous mutations in HEP-10V and four additional non-synonymous mutations in HEP-30V. Comparison among 18 recombinant HEP strains constructed by reverse genetics and vesicular stomatitis viruses pseudotyped with RABV glycoproteins indicated that the substitution P(L115H) in the phosphoprotein and G(S15R) in the glycoprotein improved viral propagation in HEP-10V, while in HEP-30V, G(V164E), G(L183P), and G(A286V) in the glycoprotein enhanced entry into Vero cells. The obtained recombinant RABV strain, rHEP-PG4 strain, with these five substitutions, is a strong candidate for production of human rabies vaccine.
Collapse
Affiliation(s)
- Michiko Harada
- Joint Graduate School of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515, Japan
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Aya Matsuu
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Eun-Sil Park
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Yusuke Inoue
- Joint Graduate School of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515, Japan
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Akihiko Uda
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Yoshihiro Kaku
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Akiko Okutani
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Guillermo Posadas-Herrera
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Keita Ishijima
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Satoshi Inoue
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Ken Maeda
- Joint Graduate School of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515, Japan.
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan.
| |
Collapse
|
3
|
Wanjari UR, Gopalakrishnan AV. Blood-testis barrier: a review on regulators in maintaining cell junction integrity between Sertoli cells. Cell Tissue Res 2024; 396:157-175. [PMID: 38564020 DOI: 10.1007/s00441-024-03894-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 03/20/2024] [Indexed: 04/04/2024]
Abstract
The blood-testis barrier (BTB) is formed adjacent to the seminiferous basement membrane. It is a distinct ultrastructure, partitioning testicular seminiferous epithelium into apical (adluminal) and basal compartments. It plays a vital role in developing and maturing spermatocytes into spermatozoa via reorganizing its structure. This enables the transportation of preleptotene spermatocytes across the BTB, from basal to adluminal compartments in the seminiferous tubules. Several bioactive peptides and biomolecules secreted by testicular cells regulate the BTB function and support spermatogenesis. These peptides activate various downstream signaling proteins and can also be the target themself, which could improve the diffusion of drugs across the BTB. The gap junction (GJ) and its coexisting junctions at the BTB maintain the immunological barrier integrity and can be the "gateway" during spermatocyte transition. These junctions are the possible route for toxicant entry, causing male reproductive dysfunction. Herein, we summarize the detailed mechanism of all the regulators playing an essential role in the maintenance of the BTB, which will help researchers to understand and find targets for drug delivery inside the testis.
Collapse
Affiliation(s)
- Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, PIN 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, PIN 632014, India.
| |
Collapse
|
4
|
Dzobo K, Dandara C. The Extracellular Matrix: Its Composition, Function, Remodeling, and Role in Tumorigenesis. Biomimetics (Basel) 2023; 8:146. [PMID: 37092398 PMCID: PMC10123695 DOI: 10.3390/biomimetics8020146] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/25/2023] Open
Abstract
The extracellular matrix (ECM) is a ubiquitous member of the body and is key to the maintenance of tissue and organ integrity. Initially thought to be a bystander in many cellular processes, the extracellular matrix has been shown to have diverse components that regulate and activate many cellular processes and ultimately influence cell phenotype. Importantly, the ECM's composition, architecture, and stiffness/elasticity influence cellular phenotypes. Under normal conditions and during development, the synthesized ECM constantly undergoes degradation and remodeling processes via the action of matrix proteases that maintain tissue homeostasis. In many pathological conditions including fibrosis and cancer, ECM synthesis, remodeling, and degradation is dysregulated, causing its integrity to be altered. Both physical and chemical cues from the ECM are sensed via receptors including integrins and play key roles in driving cellular proliferation and differentiation and in the progression of various diseases such as cancers. Advances in 'omics' technologies have seen an increase in studies focusing on bidirectional cell-matrix interactions, and here, we highlight the emerging knowledge on the role played by the ECM during normal development and in pathological conditions. This review summarizes current ECM-targeted therapies that can modify ECM tumors to overcome drug resistance and better cancer treatment.
Collapse
Affiliation(s)
- Kevin Dzobo
- Medical Research Council, SA Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
| | - Collet Dandara
- Division of Human Genetics and Institute of Infectious Disease and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
- The South African Medical Research Council-UCT Platform for Pharmacogenomics Research and Translation, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
| |
Collapse
|
5
|
Denizot AL, L'Hostis A, Sallem A, Favier S, Pierre R, Do Cruzeiro M, Guilbert T, Burlet P, Lapierre JM, Robain M, Le Lorc'H M, Vicaut E, Chatzovoulou K, Steffann J, Romana S, Méhats C, Santulli P, Patrat C, Vaiman D, Ziyyat A, Wolf JP. Cyclic fertilin-derived peptide stimulates in vitro human embryo development. F&S SCIENCE 2022; 3:49-63. [PMID: 35559995 DOI: 10.1016/j.xfss.2021.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/11/2021] [Accepted: 12/16/2021] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To study the cyclic fertilin peptide effects on preimplantation human embryogenesis. Cyclic fertilin peptide reproduces the structure of the binding site of the sperm Fertilin β (also named A Disintegrin and Metalloprotease 2: ADAM2) disintegrin domain. It binds to the oocyte membrane and increases sperm-oocyte fusion index in human and fertilization rate in mouse, providing healthy pups. It also improves human oocyte maturation and chromosome segregation in meiosis I and binds to human embryo blastomeres, suggesting that it has a membrane receptor. DESIGN Thawed human embryos at the 3 to 4 cells stage were randomly included in a dose-response study with cyclic fertilin peptide. Inner cell mass (ICM), trophectoderm (TE), and total cell numbers were evaluated in top- and good-quality blastocysts. SETTING The study was performed in an academic hospital and research laboratory. PATIENT(S) Human embryos donated for research. This project was approved by the French "Agence de la Biomédecine." INTERVENTION(S) Immunofluorescence and tissue-specific gene expression analysis, using Clariom D microarrays, were performed to study its mechanism of action. MAIN OUTCOME MEASURE(S) Cyclic fertilin peptide improves blastocyst formation by almost 20%, the concentration of 1 μM being the lowest most efficient concentration. It significantly increases twice the TE cell number, without modifying the ICM. It increases the in vitro hatching rate from 14% to 45%. RESULT(S) Cyclic fertilin peptide stimulates TE growth. In the ICM, it induces transcriptional activation of intracellular protein and vesicle-mediated transport. CONCLUSION(S) Cyclic fertilin peptide dramatically improves human embryo development potential. It could be used to supplement culture medium and improve the in vitro human embryo development. Starting supplementation immediately after fertilization, instead of day 2, could significantly upgrade assisted reproductive technology outcome.
Collapse
Affiliation(s)
- Anne-Lyse Denizot
- Team "From Gametes To Birth," Cochin Institute, Inserm U1016, CNRS UMR8104, Université de Paris, Paris, France; Department "Histologie-Embryologie-Biologie de la Reproduction," Hôpital Cochin, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Audrey L'Hostis
- Team "From Gametes To Birth," Cochin Institute, Inserm U1016, CNRS UMR8104, Université de Paris, Paris, France; Department "Histologie-Embryologie-Biologie de la Reproduction," Hôpital Cochin, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Amira Sallem
- Team "From Gametes To Birth," Cochin Institute, Inserm U1016, CNRS UMR8104, Université de Paris, Paris, France; Department "Histologie-Embryologie-Biologie de la Reproduction," Hôpital Cochin, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; Laboratoire d'Histologie-Embryologie et Cytogénétique (LR 18 ES 40), Faculté de Médecine de Monastir, Tunisie
| | - Sophie Favier
- Team "From Gametes To Birth," Cochin Institute, Inserm U1016, CNRS UMR8104, Université de Paris, Paris, France
| | - Rémi Pierre
- Homologous Recombination, Embryo Transfer and Cryopreservation Facility, Cochin Institute, University of Paris, Paris, France
| | - Marcio Do Cruzeiro
- Homologous Recombination, Embryo Transfer and Cryopreservation Facility, Cochin Institute, University of Paris, Paris, France
| | - Thomas Guilbert
- IMAG'IC facility, Cochin Institute, Inserm U1016, CNRS UMR 8104, University of Paris UMR-S1016, Paris, France
| | - Philippe Burlet
- Department "Génétique Moléculaire," Hôpital Necker-Enfants malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Jean-Michel Lapierre
- Department of "Histologie - Embryologie-Cytogénétique," Hôpital Necker-Enfants malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | | | - Marc Le Lorc'H
- Department of "Histologie - Embryologie-Cytogénétique," Hôpital Necker-Enfants malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Eric Vicaut
- Unité de Recherche Clinique, ACTION Study Group, Hôpital Fernand Widal, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Kalliopi Chatzovoulou
- Department "Génétique Moléculaire," Hôpital Necker-Enfants malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; Institut Imagine, Université de Paris, Laboratoire des Maladies Génétiques Mitochondriales. Inserm UMR1163, Paris, France
| | - Julie Steffann
- Department "Génétique Moléculaire," Hôpital Necker-Enfants malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; Institut Imagine, Université de Paris, Laboratoire des Maladies Génétiques Mitochondriales. Inserm UMR1163, Paris, France
| | - Serge Romana
- Department of "Histologie - Embryologie-Cytogénétique," Hôpital Necker-Enfants malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; Institut Imagine, Université de Paris, Laboratoire d'Embryologie et de Génétique des Malformations Congénitales, Inserm UMR1163, Paris, France
| | - Céline Méhats
- Team "From Gametes To Birth," Cochin Institute, Inserm U1016, CNRS UMR8104, Université de Paris, Paris, France
| | - Piétro Santulli
- Service de Gynécologie-Obstétrique II et de Médecine de la Reproduction, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Catherine Patrat
- Team "From Gametes To Birth," Cochin Institute, Inserm U1016, CNRS UMR8104, Université de Paris, Paris, France; Department "Histologie-Embryologie-Biologie de la Reproduction," Hôpital Cochin, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Daniel Vaiman
- Team "From Gametes To Birth," Cochin Institute, Inserm U1016, CNRS UMR8104, Université de Paris, Paris, France
| | - Ahmed Ziyyat
- Team "From Gametes To Birth," Cochin Institute, Inserm U1016, CNRS UMR8104, Université de Paris, Paris, France; Department "Histologie-Embryologie-Biologie de la Reproduction," Hôpital Cochin, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Jean Philippe Wolf
- Team "From Gametes To Birth," Cochin Institute, Inserm U1016, CNRS UMR8104, Université de Paris, Paris, France; Department "Histologie-Embryologie-Biologie de la Reproduction," Hôpital Cochin, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.
| |
Collapse
|
6
|
Abstract
Viruses are obligatory intracellular parasites that use cell proteins to take the control of the cell functions in order to accomplish their life cycle. Studying the viral-host interactions would increase our knowledge of the viral biology and mechanisms of pathogenesis. Studies on pathogenesis mechanisms of lyssaviruses, which are the causative agents of rabies, have revealed some important host protein partners for viral proteins, especially for most studied species, i.e. RABV. In this review article, the key physical lyssavirus-host protein interactions, their contributions to rabies infection, and their exploitation are discussed to improve the knowledge about rabies pathogenesis.
Collapse
|
7
|
Pan K, Xie Y. LncRNA FOXC2-AS1 enhances FOXC2 mRNA stability to promote colorectal cancer progression via activation of Ca 2+-FAK signal pathway. Cell Death Dis 2020; 11:434. [PMID: 32513911 PMCID: PMC7280533 DOI: 10.1038/s41419-020-2633-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 12/18/2022]
Abstract
Long noncoding RNAs (lncRNAs) have been confirmed, which are involved in tumorigenesis and metastasis in colorectal cancer (CRC). FOXC2 antisense RNA 1 (FOXC2-AS1) was reported, facilitating the proliferation and progression in several cancers. However, the role of FOXC2-AS1 in CRC cell migration and metastasis is not unclear. In this study, we observed that lncRNA FOXC2-AS1 was upregulated in CRC tissues, and its high expression indicated the poor survival in CRC patients. Meanwhile, FOXC2-AS1 was higher in CRC tissues with metastasis than that of nonmetastatic tumor tissues. We found that FOXC2-AS1 was predominately expressed in the nucleus of tissues and cells. FOXC2-AS1 knockdown suppressed CRC cell growth, invasion, and metastasis in vitro and in vivo. Moreover, FOXC2-AS1 could positively regulate the neighboring gene FOXC2 and stabilized FOXC2 mRNA by forming a RNA duplex. Meanwhile, ectopic expression of FOXC2 could obviously alleviate the suppressed effects caused by silencing FOXC2-AS1. For the mechanism, FOXC2-AS1 knockdown could reduce intracellular Ca2+ levels, inhibited FA formation and FAK signaling, and these suppressed effects were mitigated by increasing FOXC2 expression. These results demonstrated that FOXC2-AS1 enhances FOXC2 mRNA stability to promote CRC proliferation, migration, and invasion by activation of Ca2+-FAK signaling, which implicates that FOXC2-AS1 may represent a latent effective therapeutic target for CRC progression.
Collapse
Affiliation(s)
- Ke Pan
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yong Xie
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
8
|
Song J, Ye B, Liu H, Bi R, Zhang N, Hu J, Luo E. Fak-Mapk, Hippo and Wnt signalling pathway expression and regulation in distraction osteogenesis. Cell Prolif 2018; 51:e12453. [PMID: 29504176 PMCID: PMC6528869 DOI: 10.1111/cpr.12453] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 01/27/2018] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES To investigate the mechanism of mechanical stimulation in bone formation and regeneration during distraction osteogenesis. MATERIALS AND METHODS In this study, microarray technology was used to investigate the time course of bone-related molecular changes in distraction osteogenesis in rats. Real-time PCR and Western-blot analyses were used to confirm the expression of genes identified in microarrays. Meanwhile, we used a lentivirus vector to inhibit Fak expression, in order to identify the osteogenic effect of Fak and Fak-Mapk pathway during distraction osteogenesis. RESULTS Several components of the Wnt and Hippo pathways were found to be up- or down-regulated during distraction osteogenesis by microarray. Meanwhile, it was found that Fak, Src, Raf-1, Erk1, Jnk and p38-Mapk were up-regulated during gradual distraction, compared with consolidation. To further determine whether Fak-Mapk pathway played an important role in distraction osteogenesis, Fak was disrupted with a lentivirus vector. The expressions levels of p-Fak, p-Erk1/2, p-JNK and p-p38Mapk were decreased. Meanwhile, a poor early and late osteogenesis effect was found in the shRNA-Fak group. CONCLUSION It was inferred that the mechanical stimulus induces increased expression of Fak and activates Fak-Mapk pathway, by activation of Erk, Jnk and p38-Mapk pathway, and that Fak at least, in part, plays an important role in maintaining osteogenic effect by activating Fak-Mapk pathway during distraction osteogenesis.
Collapse
Affiliation(s)
- Jian Song
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesDepartment of oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Bin Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesDepartment of oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesDepartment of oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Ruiye Bi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesDepartment of oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Nian Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesDepartment of oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Jing Hu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesDepartment of oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - En Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesDepartment of oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
9
|
Rajshankar D, Wang Y, McCulloch CA. Osteogenesis requires FAK-dependent collagen synthesis by fibroblasts and osteoblasts. FASEB J 2016; 31:937-953. [PMID: 27881487 DOI: 10.1096/fj.201600645r] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 11/14/2016] [Indexed: 11/11/2022]
Abstract
Focal adhesion kinase (FAK) is critical in adhesion-dependent signaling, but its role in osteogenesis in vivo is ill defined. We deleted Fak in fibroblasts and osteoblasts in Floxed-Fak mice bred with those expressing Cre-recombinase driven by 3.6-kb α1(I)-collagen promoter. Compared with wild-type (WT), conditional FAK-knockout (CFKO) mice were shorter (2-fold; P < 0.0001) and had crooked, shorter tails (50%; P < 0.0001). Microcomputed tomography analysis showed reduced bone volume (4-fold in tails; P < 0.0001; 2-fold in mandibles; P < 0.0001), whereas bone surface area/bone volume increased (3-fold in tails; P < 0.0001; 2.5-fold in mandibles; P < 0.001). Collagen density and fiber alignment in periodontal ligament were reduced by 4-fold (P < 0.0001) and 30% (P < 0.05), respectively, in CFKO mice. In cultured CFKO osteoblasts, mineralization at d 7 and mineralizing colony-forming units at d 21 were 30% (P < 0.0001) and >3-fold less than WT, respectively. Disruptions of FAK function in osteoblasts by conditional knockout, siRNA-knockdown, or FAK inhibitor reduced mRNA and protein expression of Runx2 (>30%), Osterix (>25%), and collagen-1 (2-fold). Collagen synthesis was abrogated in WT osteoblasts with Runx2 knockdown and in Fak-null fibroblasts transfected with an FAK kinase domain mutant or a kinase-impaired mutant (Y397F). These data indicate that FAK regulates osteogenesis through transcription factors that regulate collagen synthesis.-Rajshankar, D., Wang, Y., McCulloch, C. A. Osteogenesis requires FAK-dependent collagen synthesis by fibroblasts and osteoblasts.
Collapse
Affiliation(s)
- Dhaarmini Rajshankar
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Yongqiang Wang
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Christopher A McCulloch
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Fakhry CT, Choudhary P, Gutteridge A, Sidders B, Chen P, Ziemek D, Zarringhalam K. Interpreting transcriptional changes using causal graphs: new methods and their practical utility on public networks. BMC Bioinformatics 2016; 17:318. [PMID: 27553489 PMCID: PMC4995651 DOI: 10.1186/s12859-016-1181-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 08/11/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Inference of active regulatory cascades under specific molecular and environmental perturbations is a recurring task in transcriptional data analysis. Commercial tools based on large, manually curated networks of causal relationships offering such functionality have been used in thousands of articles in the biomedical literature. The adoption and extension of such methods in the academic community has been hampered by the lack of freely available, efficient algorithms and an accompanying demonstration of their applicability using current public networks. RESULTS In this article, we propose a new statistical method that will infer likely upstream regulators based on observed patterns of up- and down-regulated transcripts. The method is suitable for use with public interaction networks with a mix of signed and unsigned causal edges. It subsumes and extends two previously published approaches and we provide a novel algorithmic method for efficient statistical inference. Notably, we demonstrate the feasibility of using the approach to generate biological insights given current public networks in the context of controlled in-vitro overexpression experiments, stem-cell differentiation data and animal disease models. We also provide an efficient implementation of our method in the R package QuaternaryProd available to download from Bioconductor. CONCLUSIONS In this work, we have closed an important gap in utilizing causal networks to analyze differentially expressed genes. Our proposed Quaternary test statistic incorporates all available evidence on the potential relevance of an upstream regulator. The new approach broadens the use of these types of statistics for highly curated signed networks in which ambiguities arise but also enables the use of networks with unsigned edges. We design and implement a novel computational method that can efficiently estimate p-values for upstream regulators in current biological settings. We demonstrate the ready applicability of the implemented method to analyze differentially expressed genes using the publicly available networks.
Collapse
Affiliation(s)
- Carl Tony Fakhry
- Department of Computer Science, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, 02125, USA
| | - Parul Choudhary
- Computational Sciences, Pfizer Worldwide Research & Development, Cambridge, USA
| | - Alex Gutteridge
- Computational Sciences, Pfizer Worldwide Research & Development, Cambridge, USA
| | - Ben Sidders
- Computational Sciences, Pfizer Worldwide Research & Development, Cambridge, USA
| | - Ping Chen
- Department of Engineering, University of Massachusetts Boston, Boston, 100 Morrissey Boulevard02125, USA
| | - Daniel Ziemek
- Computational Sciences, Pfizer Worldwide Research & Development, Berlin, USA
| | - Kourosh Zarringhalam
- Department of Mathematics, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, 02125, USA.
| |
Collapse
|
11
|
Shortrede JE, Uzair ID, Neira FJ, Flamini MI, Sanchez AM. Paxillin, a novel controller in the signaling of estrogen to FAK/N-WASP/Arp2/3 complex in breast cancer cells. Mol Cell Endocrinol 2016; 430:56-67. [PMID: 27095481 DOI: 10.1016/j.mce.2016.04.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 04/14/2016] [Accepted: 04/15/2016] [Indexed: 11/25/2022]
Abstract
Breast cancer is the major cause of cancer-related death in women. Its treatment is particularly difficult when metastasis occurs. The ability of cancer cells to move and invade the surrounding environment is the basis of local and distant metastasis. Cancer cells are able to remodel the actin cytoskeleton, which requires the recruitment of numerous structural and regulatory proteins that modulate actin filaments dynamics, including Paxillin or the Neural Wiskott-Aldrich Syndrome Protein (N-WASP). We show that 17-β estradiol (E2) induces phosphorylation of Paxillin and its translocation toward membrane sites where focal adhesion complexes are assembled. This cascade is triggered by a Gαi1/Gβ protein-dependent signaling of estrogen receptor α (ERα) to c-Src, focal adhesion kinase (FAK) and Paxillin. Within this complex, activated Paxillin recruits the small GTPase Cdc42, which triggers N-WASP phosphorylation. This results in the redistribution of Arp2/3 complexes at sites where membrane structures related to cell movement are formed. Recruitment of Paxillin, Cdc42 and N-WASP is necessary for cell adhesion, migration and invasion induced by E2 in breast cancer cells. In parallel, we investigated whether Raloxifene (RAL), a selective estrogen receptor modulator (SERMs), could inhibit or revert the effects of E2 in breast cancer cell movement. We found that, in the presence of E2, RAL acts as an ER antagonist and displays an inhibitory effect on estrogen-promoted cell adhesion and migration via FAK/Paxillin/N-WASP. Our findings identify an original mechanism through which estrogen regulates breast cancer cell motility and invasion via Paxillin. These results may have clinical relevance for the development of new therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Jorge Eduardo Shortrede
- Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Research Council of Argentina, Mendoza, Argentina
| | - Ivonne Denise Uzair
- Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Research Council of Argentina, Mendoza, Argentina
| | - Flavia Judith Neira
- Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Research Council of Argentina, Mendoza, Argentina
| | - Marina Inés Flamini
- Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Research Council of Argentina, Mendoza, Argentina
| | - Angel Matías Sanchez
- Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Research Council of Argentina, Mendoza, Argentina.
| |
Collapse
|
12
|
Biological basis of distraction osteogenesis – A review. JOURNAL OF ORAL AND MAXILLOFACIAL SURGERY MEDICINE AND PATHOLOGY 2016. [DOI: 10.1016/j.ajoms.2015.05.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
13
|
Sawicka KM, Seeliger M, Musaev T, Macri LK, Clark RA. Fibronectin Interaction and Enhancement of Growth Factors: Importance for Wound Healing. Adv Wound Care (New Rochelle) 2015; 4:469-478. [PMID: 26244103 DOI: 10.1089/wound.2014.0616] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/30/2015] [Indexed: 12/21/2022] Open
Abstract
Significance: This critical review focuses on interactions between cells, fibronectin (FN), and growth factors (GF). Recent Advances: Initially, the extracellular matrix (ECM) was thought to serve simply as a reservoir for GFs that would be released as soluble ligands during proteolytic degradation of ECM. This view was rather quickly extended by the observation that ECM could concentrate GFs to the pericellular matrix for more efficient presentation to cell surface receptors. However, recent reports support much more complex interactions among GFs and ECM molecules, particularly FN, and the way these interactions can fine-tune cell responses to the microenvironment. Critical Issues: Wounds that are unable to synthesize and sustain a functional ECM cannot optimally benefit from endogenous or exogenous GFs. Therefore, GF treatments have recently focused on utilizing ECM molecules as delivery vehicles. Thus, ECM can influence GF stability and activity, and GFs can modulate the ECM activity. Hence, both individually and together, ECM and GFs modulate cells that in turn control the type and level of GFs and ECM in the pericellular environment that ultimately results in new tissue generation. Although many ECM components are important for optimal tissue regeneration and wound healing, FN stands out as absolutely critical not only for wound healing and tissue regeneration but also for embryogenesis and morphogenesis. Future Directions: Understanding ECM/GF interactions will greatly facilitate our understanding of normal wound repair and regeneration, the failure of wounds to heal, and how the latter can be salvaged with proper ECM/GF combinations.
Collapse
Affiliation(s)
- Katarzyna M. Sawicka
- Department of Dermatology, Stony Brook School of Medicine, Stony Brook, New York
| | - Markus Seeliger
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
| | - Tagai Musaev
- Albert Einstein College of Medicine, New York City, New York
| | - Lauren K. Macri
- New Jersey Center for Biomaterials Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Richard A.F. Clark
- Department of Dermatology, Stony Brook School of Medicine, Stony Brook, New York
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York
| |
Collapse
|
14
|
Selective targeting of FAK–Pyk2 axis by alpha-naphthoflavone abrogates doxorubicin resistance in breast cancer cells. Cancer Lett 2015; 362:25-35. [DOI: 10.1016/j.canlet.2015.03.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/05/2015] [Accepted: 03/09/2015] [Indexed: 01/26/2023]
|
15
|
Rutkunas V, Bukelskiene V, Sabaliauskas V, Balciunas E, Malinauskas M, Baltriukiene D. Assessment of human gingival fibroblast interaction with dental implant abutment materials. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2015; 26:169. [PMID: 25804303 DOI: 10.1007/s10856-015-5481-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Accepted: 02/09/2015] [Indexed: 06/04/2023]
Abstract
The biocompatibility of dental implant abutment materials depends on numerous factors including the nature of the material, its chemical composition, roughness, texture, hydrophilicity and surface charge. The aim of the present study was to compare the viability and adhesion strength of human gingival fibroblasts (HGFs) grown on several dental materials used in implant prosthodontics. Surfaces of the tested materials were assessed using an optical imaging profiler. For material toxicity and cellular adhesion evaluation, primary human gingival fibroblast cells were used. To evaluate the strength of cellular adhesion, gingival fibroblasts were cultured on the tested materials and subjected to lateral shear forces by applying 300 and 500 rpm shaking intensities. Focal adhesion kinase (FAK) expression and phosphorylation in cells grown on the specimens were registered by cell-based ELISA. There was a tendency of fibroblast adhesion strength to decrease in the following order: sandblasted titanium, polished titanium, sandblasted zirconium oxide, polished zirconium oxide, gold-alloy, chrome-cobalt alloy. Higher levels of total as well as phospho-FAK protein were registered in HGFs grown on roughened titanium. Material type and surface processing technique have an impact on gingival fibroblast interaction with dental implant abutment materials.
Collapse
Affiliation(s)
- Vygandas Rutkunas
- Department of Prosthodontics, Institute of Odontology, Faculty of Medicine, Vilnius University, Zalgirio str. 115, 08217, Vilnius, Lithuania,
| | | | | | | | | | | |
Collapse
|
16
|
Focal adhesion kinase is involved in rabies virus infection through its interaction with viral phosphoprotein P. J Virol 2014; 89:1640-51. [PMID: 25410852 DOI: 10.1128/jvi.02602-14] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
UNLABELLED The rabies virus (RABV) phosphoprotein P is a multifunctional protein: it plays an essential role in viral transcription and replication, and in addition, RABV P has been identified as an interferon antagonist. Here, a yeast two-hybrid screen revealed that RABV P interacts with the focal adhesion kinase (FAK). The binding involved the 106-to-131 domain, corresponding to the dimerization domain of P and the C-terminal domain of FAK containing the proline-rich domains PRR2 and PRR3. The P-FAK interaction was confirmed in infected cells by coimmunoprecipitation and colocalization of FAK with P in Negri bodies. By alanine scanning, we identified a single mutation in the P protein that abolishes this interaction. The mutant virus containing a substitution of Ala for Arg in position 109 in P (P.R109A), which did not interact with FAK, is affected at a posttranscriptional step involving protein synthesis and viral RNA replication. Furthermore, FAK depletion inhibited viral protein expression in infected cells. This provides the first evidence of an interaction of RABV with FAK that positively regulates infection. IMPORTANCE Rabies virus exhibits a small genome that encodes a limited number of viral proteins. To maintain efficient virus replication, some of them are multifunctional, such as the phosphoprotein P. We and others have shown that P establishes complex networks of interactions with host cell components. These interactions have revealed much about the role of P and about host-pathogen interactions in infected cells. Here, we identified another cellular partner of P, the focal adhesion kinase (FAK). Our data shed light on the implication of FAK in RABV infection and provide evidence that P-FAK interaction has a proviral function.
Collapse
|
17
|
Li Y, Moretto-Zita M, Leon-Garcia S, Parast MM. p63 inhibits extravillous trophoblast migration and maintains cells in a cytotrophoblast stem cell-like state. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:3332-43. [PMID: 25307348 DOI: 10.1016/j.ajpath.2014.08.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 08/05/2014] [Accepted: 08/14/2014] [Indexed: 11/16/2022]
Abstract
Proper differentiation of placental epithelial cells, called trophoblast, is required for implantation. Early during placentation, trophoblast cell columns help anchor the developing embryo in the uterine wall. Although proximally continuous with villous cytotrophoblast (CTB) distally, these cells differentiate into invasive extravillous trophoblast. We previously reported that p63, a p53 family member, is highly expressed in proliferative villous CTB and required for induction of the trophoblast lineage in human pluripotent stem cells. We now further explore its function in human trophoblast by using both primary CTB from the early placenta and established trophoblast cell lines. We show that p63 is expressed in epidermal growth factor receptor-positive CTB and that its expression decreases with differentiation into HLA-G(+) extravillous trophoblast. In trophoblast cell lines, p63 is expressed in JEG3 cells but absent from HTR8 cells. Overexpression of p63 in both cell lines enhances cell proliferation and significantly reduces cell migration; conversely, down-regulation of p63 in JEG3 cells reduces cell proliferation and restores cell migration. Analysis of epithelial-to-mesenchymal transition, cell adhesion, and matrix degradation pathways shows that p63 blocks epithelial-to-mesenchymal transition, promotes a CTB-specific cell adhesion profile, and inhibits expression of matrix metalloproteinases. Taken together, these data show that p63 maintains the proliferative CTB state, at least partially through regulation of epithelial-to-mesenchymal transition, cell adhesion, and matrix degradation pathways.
Collapse
Affiliation(s)
- Yingchun Li
- Department of Pathology, University of California San Diego, La Jolla, California; Department of Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Matteo Moretto-Zita
- Department of Pathology, University of California San Diego, La Jolla, California; Department of Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Sandra Leon-Garcia
- Department of Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California; Department of Reproductive Medicine, University of California San Diego, La Jolla, California
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, California; Department of Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California.
| |
Collapse
|
18
|
Neogenin as a receptor for early cell fate determination in preimplantation mouse embryos. PLoS One 2014; 9:e101989. [PMID: 25013897 PMCID: PMC4094428 DOI: 10.1371/journal.pone.0101989] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 06/12/2014] [Indexed: 01/03/2023] Open
Abstract
The first cell lineage determination in embryos takes place when two cell populations are set apart, each differentiating into the trophectoderm (TE) and inner cell mass (ICM), respectively. It is widely believed that position/polarity cues play a key role in triggering this differentiation, but it remains unclear how extracellular cues are transduced into cell fate determination. Here, we provide evidence that supports that neogenin is implicated in relaying extracellular cues into the first cell fate determination in preimplantation mouse embryos. A polarized and transient distribution of neogenin was manifested in early blastomeres. Neogenin up-regulation by its overexpression accelerated ICM development in the blastocyst concomitant with the activation of the ICM-specific transcription factors Oct3/4, Sox2, and Nanog while its depletion by small hairpin RNAs (shRNAs) caused a developmental abnormality of poorly endowed ICM accompanied by the deactivation of Oct3/4, Sox2, and Nanog. Treatment with netrin-1 among neogenin ligands further impaired both embryonic development and ICM formation while repulsive guidance molecule c (RGMc) led to opposite consequences, enhancing ICM formation. From this study, we propose a model whereby neogenin interprets its own expression level to control the first cell fate determination in response to extracellular cues.
Collapse
|
19
|
Zhu J, Clark RAF. Fibronectin at select sites binds multiple growth factors and enhances their activity: expansion of the collaborative ECM-GF paradigm. J Invest Dermatol 2014; 134:895-901. [PMID: 24335899 PMCID: PMC3961531 DOI: 10.1038/jid.2013.484] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 10/10/2013] [Accepted: 10/25/2013] [Indexed: 01/23/2023]
Abstract
Intensive research has demonstrated that extracellular matrix (ECM) molecules and growth factors (GF) collaborate at many different levels. The ability of ECM to modulate GF signals has important implications in tissue formation and homeostasis as well as novel therapies for acute and chronic wounds. Recently, a number of GF-binding sites was identified in fibronectin (FN) and was shown to provide another layer of regulation on GF signaling. Here, we review these new findings on FN interaction with GF in the context of general ways ECM molecules regulate GF signaling.
Collapse
Affiliation(s)
- Jia Zhu
- Department of Biochemistry, Stony Brook University, Stony Brook, New York, USA
| | - Richard A F Clark
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA; Department of Dermatology, Stony Brook University, Stony Brook, New York, USA; Department of Medicine, Stony Brook University, Stony Brook, New York, USA.
| |
Collapse
|
20
|
Jo A, Park H, Lee SH, Ahn SH, Kim HJ, Park EM, Choi YH. SHP-2 binds to caveolin-1 and regulates Src activity via competitive inhibition of CSK in response to H2O2 in astrocytes. PLoS One 2014; 9:e91582. [PMID: 24632723 PMCID: PMC3954793 DOI: 10.1371/journal.pone.0091582] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 02/12/2014] [Indexed: 01/26/2023] Open
Abstract
Reactive oxygen species (ROS) regulate diverse cellular functions by triggering signal transduction events, such as Src and mitogen-activated protein (MAP) kinases. Here, we report the role of caveolin-1 and Src homology 2 domain-containing protein tyrosine phosphatase 2 (SHP-2) in H2O2-induced signaling pathway in brain astrocytes. H2O2-mediated oxidative stress induced phosphorylation of caveolin-1 and association between p-caveolin-1 and SHP-2. SHP-2 specifically bound to wild-type caveolin-1 similarly to c-Src tyrosine kinase (CSK), but not to phosphorylation-deficient mutant of caveolin-1 (Y14A), and interfered with complex formation between caveolin-1 and CSK. In the presence of CSK siRNA, binding between caveolin-1 and SHP-2 was enhanced by H2O2 treatment, which led to reduced Src phosphorylation at tyrosine (Tyr) 530 and enhanced Src phosphorylation at Tyr 419. In contrast, siRNA targeting of SHP-2 facilitated H2O2-mediated interaction between caveolin-1 and CSK and enhanced Src phosphorylation at Tyr 530, leading to subsequent decrease in Src downstream signaling, such as focal adhesion kinase (FAK) and extracellular signal-related kinase (ERK). Our results collectively indicate that SHP-2 alters Src kinase activity by interfering with the complex formation between CSK and phosphotyrosine caveolin-1 in the presence of H2O2, thus functions as a positive regulator in Src signaling under oxidative stress in brain astrocytes.
Collapse
Affiliation(s)
- Ara Jo
- Department of Physiology, Tissue Injury Defense Research Center, Ewha Womans University School of Medicine, Seoul, Korea
| | - Hyunju Park
- Department of Physiology, Tissue Injury Defense Research Center, Ewha Womans University School of Medicine, Seoul, Korea
| | - Sung-Hee Lee
- Department of Physiology, Tissue Injury Defense Research Center, Ewha Womans University School of Medicine, Seoul, Korea
| | - So-Hee Ahn
- Department of Physiology, Tissue Injury Defense Research Center, Ewha Womans University School of Medicine, Seoul, Korea
| | - Hee Ja Kim
- Department of Physiology, Tissue Injury Defense Research Center, Ewha Womans University School of Medicine, Seoul, Korea
| | - Eun-Mi Park
- Department of Pharmacology, Tissue Injury Defense Research Center, Ewha Womans University School of Medicine, Seoul, Korea
| | - Youn-Hee Choi
- Department of Physiology, Tissue Injury Defense Research Center, Ewha Womans University School of Medicine, Seoul, Korea
| |
Collapse
|
21
|
Abstract
Tumorigenesis is the process by which normal cells evolve the capacity to evade and overcome the constraints usually placed upon their growth and survival. To ensure the integrity of organs and tissues, the balance of cell proliferation and cell death is tightly maintained. The proteins controlling this balance are either considered oncogenes, which promote tumorigenesis, or tumor suppressors, which prevent tumorigenesis. Phosphoinositide 3-kinase enhancer (PIKE) is a family of GTP-binding proteins that possess anti-apoptotic functions and play an important role in the central nervous system. Notably, accumulating evidence suggests that PIKE is a proto-oncogene involved in tumor progression. The PIKE gene (CENTG1) is amplified in a variety of human cancers, leading to the resistance against apoptosis and the enhancement of invasion. In this review, we will summarize the functions of PIKE proteins in tumorigenesis and discuss their potential implications in cancer therapy.
Collapse
|
22
|
Inhibition of retinal ganglion cell axonal outgrowth through the Amino-Nogo-A signaling pathway. Neurochem Res 2013; 38:1365-74. [PMID: 23579387 DOI: 10.1007/s11064-013-1032-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 03/03/2013] [Accepted: 03/26/2013] [Indexed: 01/10/2023]
Abstract
Nogo-A is a myelin-derived inhibitor playing a pivotal role in the prevention of axonal regeneration. A functional domain of Nogo-A, Amino-Nogo, exerts an inhibitory effect on axonal regeneration, although the mechanism is unclear. The present study investigated the role of the Amino-Nogo-integrin signaling pathway in primary retinal ganglion cells (RGCs) with respect to axonal outgrowth, which is required for axonal regeneration. Immunohistochemistry showed that integrin αv, integrin α5 and FAK were widely expressed in the visual system. Thy-1 and GAP-43 immunofluorescence showed that axonal outgrowth of RGCs was promoted by Nogo-A siRNA and a peptide antagonist of the Nogo-66 functional domain of Nogo-A (Nep1-40), and inhibited by a recombinant rat Nogo-A-Fc chimeric protein (Δ20). Western blotting revealed increased integrin αv and p-FAK expression in Nogo-A siRNA group, decreased integrin αv expression in Δ20 group and decreased p-FAK expression in Nep1-40 group. Integrin α5 expression was not changed in any group. RhoA G-LISA showed that RhoA activation was inhibited by Nogo-A siRNA and Δ20, but increased by Nep1-40 treatment. These results suggest that Amino-Nogo inhibits RGC axonal outgrowth primarily through the integrin αv signaling pathway.
Collapse
|
23
|
Gualandi C, Govoni M, Foroni L, Valente S, Bianchi M, Giordano E, Pasquinelli G, Biscarini F, Focarete ML. Ethanol disinfection affects physical properties and cell response of electrospun poly(l-lactic acid) scaffolds. Eur Polym J 2012. [DOI: 10.1016/j.eurpolymj.2012.09.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
24
|
Matsumoto M, Tamura M, Miyamoto T, Furuno Y, Kabashima N, Serino R, Shibata T, Kanegae K, Takeuchi M, Abe H, Okazaki M, Otsuji Y. Impacts of icodextrin on integrin-mediated wound healing of peritoneal mesothelial cells. Life Sci 2012; 90:917-23. [DOI: 10.1016/j.lfs.2012.04.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 03/28/2012] [Accepted: 04/16/2012] [Indexed: 10/28/2022]
|
25
|
Abstract
The blood-testis barrier (BTB) is one of the tightest blood-tissue barriers in the mammalian body. It divides the seminiferous epithelium into the basal and the apical (adluminal) compartments. Meiosis I and II, spermiogenesis, and spermiation all take place in a specialized microenvironment behind the BTB in the apical compartment, but spermatogonial renewal and differentiation and cell cycle progression up to the preleptotene spermatocyte stage take place outside of the BTB in the basal compartment of the epithelium. However, the BTB is not a static ultrastructure. Instead, it undergoes extensive restructuring during the seminiferous epithelial cycle of spermatogenesis at stage VIII to allow the transit of preleptotene spermatocytes at the BTB. Yet the immunological barrier conferred by the BTB cannot be compromised, even transiently, during the epithelial cycle to avoid the production of antibodies against meiotic and postmeiotic germ cells. Studies have demonstrated that some unlikely partners, namely adhesion protein complexes (e.g., occludin-ZO-1, N-cadherin-β-catenin, claudin-5-ZO-1), steroids (e.g., testosterone, estradiol-17β), nonreceptor protein kinases (e.g., focal adhesion kinase, c-Src, c-Yes), polarity proteins (e.g., PAR6, Cdc42, 14-3-3), endocytic vesicle proteins (e.g., clathrin, caveolin, dynamin 2), and actin regulatory proteins (e.g., Eps8, Arp2/3 complex), are working together, apparently under the overall influence of cytokines (e.g., transforming growth factor-β3, tumor necrosis factor-α, interleukin-1α). In short, a "new" BTB is created behind spermatocytes in transit while the "old" BTB above transiting cells undergoes timely degeneration, so that the immunological barrier can be maintained while spermatocytes are traversing the BTB. We also discuss recent findings regarding the molecular mechanisms by which environmental toxicants (e.g., cadmium, bisphenol A) induce testicular injury via their initial actions at the BTB to elicit subsequent damage to germ-cell adhesion, thereby leading to germ-cell loss, reduced sperm count, and male infertility or subfertility. Moreover, we also critically evaluate findings in the field regarding studies on drug transporters in the testis and discuss how these influx and efflux pumps regulate the entry of potential nonhormonal male contraceptives to the apical compartment to exert their effects. Collectively, these findings illustrate multiple potential targets are present at the BTB for innovative contraceptive development and for better delivery of drugs to alleviate toxicant-induced reproductive dysfunction in men.
Collapse
Affiliation(s)
- C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, USA.
| | | |
Collapse
|
26
|
Xu B, Song G, Ju Y. Effect of focal adhesion kinase on the regulation of realignment and tenogenic differentiation of human mesenchymal stem cells by mechanical stretch. Connect Tissue Res 2011; 52:373-9. [PMID: 21401419 DOI: 10.3109/03008207.2010.541961] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Focal adhesion kinase (FAK) is a focal adhesion-associated protein kinase involved in cell adhesion and spreading. It is recruited as a participant in focal adhesion dynamics between cells and has a role in cell motility, differentiation, and survival. The role of FAK in the differentiation of human mesenchymal stem cells (hMSCs), however, is not well understood, particularly in terms of tenogenic differentiation. In this study, we reported that FAK regulates the mechanical stretch-induced realignment of hMSCs. We showed that FAK can be activated by mechanical stretch and, with a 10 μM PF 573228 (a novel small molecule inhibitor of FAK) treatment, FAK autophosphorylation at Tyr397 is significantly decreased. Moreover, our findings demonstrated that this decrease in FAK autophosphorylation at Tyr397 leads to the attenuation of upregulation of mechanical stretch-induced mRNA expression of tendon-related genes, including type I collagen, type III collagen, tenascin-C, and scleraxis. These results indicate that the FAK signaling molecule plays an important role in regulating cell realignment and tenogenic differentiation of hMSCs when induced by mechanical stretch. Collectively, our findings provide novel insight into the role of FAK in the realignment and mechanotransduction of hMSCs during the process of tenogenic differentiation induced by mechanical stretch.
Collapse
Affiliation(s)
- Baiyao Xu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, China
| | | | | |
Collapse
|
27
|
Lie PPY, Cheng CY, Mruk DD. The biology of the desmosome-like junction a versatile anchoring junction and signal transducer in the seminiferous epithelium. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2011; 286:223-69. [PMID: 21199783 PMCID: PMC4381909 DOI: 10.1016/b978-0-12-385859-7.00005-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mammalian spermatogenesis, a complex process that involves the movement of developing germ cells across the seminiferous epithelium, entails extensive restructuring of Sertoli-Sertoli and Sertoli-germ cell junctions. Presently, it is not entirely clear how zygotene spermatocytes gain entry into the adluminal compartment of the seminiferous epithelium, which is sealed off from the systemic circulation by the Sertoli cell component of the blood-testis barrier, without compromising barrier integrity. To begin to address this question, it is critical that we first have a good understanding of the biology and the regulation of different types of Sertoli-Sertoli and Sertoli-germ cell junctions in the testis. Supported by recent studies in the field, we discuss how crosstalk between different types of junctions contributes to their restructuring during germ cell movement across the blood-testis barrier. We place special emphasis on the emerging role of desmosome-like junctions as signal transducers during germ cell movement across the seminiferous epithelium.
Collapse
Affiliation(s)
- Pearl P Y Lie
- Population Council, Center for Biomedical Research, New York, New York, USA
| | | | | |
Collapse
|
28
|
Lee J, Borboa AK, Chun HB, Baird A, Eliceiri BP. Conditional deletion of the focal adhesion kinase FAK alters remodeling of the blood-brain barrier in glioma. Cancer Res 2010; 70:10131-40. [PMID: 21159635 PMCID: PMC3059220 DOI: 10.1158/0008-5472.can-10-2740] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Gliomas generally infiltrate the surrounding normal brain parenchyma, a process associated with increased vascular permeability (VP) and dysregulation of the blood-brain barrier (BBB). However, the molecular mechanisms underlying glioma-induced VP in the brain remain poorly understood. Using a conditional, endothelium-specific deletion of the focal adhesion kinase (FAK) in the mouse (FAK CKO), we show that FAK is critical for destabilization of the tumor endothelium in tumor-bearing mice, with mutant mice exhibiting a relatively normalized vasculature compared with wild-type mice (FAK WT). Tumor vessels in the FAK CKO mice displayed reduced VP compared with FAK WT mice, resulting in reduced tumor growth. Additionally, FAK CKO mice displayed partial restoration of cell-cell junction proteins in the tumor vessels and astrocyte-endothelium interactions in tumors, revealing an additional role of astrocytes in mediating tumor-induced VP. Together, these results provide genetic evidence that FAK is a mediator of tumor-induced VP in the brain. Our findings may help understand how therapeutics might be used to regulate specific cell-type interactions to restore BBB structure/function in cancer and perhaps other pathologic conditions.
Collapse
Affiliation(s)
- Jisook Lee
- Department of Surgery, University of California San Diego, California 92103-8236, USA
| | | | | | | | | |
Collapse
|
29
|
Huang AL, Wan Y, Liao DY, Hu HZ, Wei L, Wang XH, Wen YJ, Li J, Chen LJ, Kan B, Chen P, Wang YS, Chen X, Zhao X, Deng HX, Wei YQ. Suppression of human MDA-MB-435S tumor by U6 promoter-driven short hairpin RNAs targeting focal adhesion kinase. J Cancer Res Clin Oncol 2010; 136:1229-42. [PMID: 20140743 DOI: 10.1007/s00432-010-0773-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2008] [Accepted: 01/14/2010] [Indexed: 02/05/2023]
Abstract
PURPOSE Focal adhesion kinase (FAK) is a non-receptor protein tyrosine kinase implicated in cancer cell survival, proliferation, and in various steps in the metastatic cascade. In the present study, we took advantage of a cationic liposome as gene carrier and targeted FAK function through both in vitro and in vivo approaches. METHODS We utilized a plasmid-encoded hairpin RNA targeting the human FAK mRNA (pGensil2-shRNA/FAK), as a means to inhibit FAK expression for evaluating its anti-tumor effect in vitro and in vivo. Human MDA-MB-435S breast cancer cells were transfected with pGensil2-shRNA/FAK and examined for apoptosis by propidium iodide staining, DNA ladder, and flow cytometric analysis. For in vivo study, subcutaneous breast carcinomatosis models in nude mice were established to evaluate the therapeutic potential of pGensil2-shRNA/FAK. Assessments of proliferation (Ki-67), apoptosis (TUNEL) and angiogenesis (CD31) were done using immunohistochemical analysis. RESULTS Transcripts expressed from plasmid both in vitro and in vivo were identified by northern blot analysis. pGensil2-shRNA/FAK effectively down-regulated the expression of FAK as demonstrated in vitro by real time RT-PCR and western blot analysis, whereas by real time RT-PCR and IHC staining of MDA-MB-435S tumors growing subcutaneously. Breast cancer cells lacking FAK expression undergo apoptosis in vitro. Systemic delivery of cationic liposome-complexed plasmids targeting FAK, resulted in the diminishment of subcutaneous tumor growth beyond the effects observed with liposomes carrying a non-specific shRNA. This diminishment in growth was associated with elevated levels of apoptosis (TUNEL staining), decreased cell proliferation (Ki-67 staining) and diminished endothelial cell density (CD31 staining). CONCLUSION These results indicate that the systemic delivery of plasmid DNA targeting FAK function using cationic liposome as a gene carrier, represents a promising avenue for breast cancer therapy.
Collapse
Affiliation(s)
- An-liang Huang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Biological role of dystroglycan in Schwann cell function and its implications in peripheral nervous system diseases. J Biomed Biotechnol 2010; 2010:740403. [PMID: 20625412 PMCID: PMC2896880 DOI: 10.1155/2010/740403] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Accepted: 04/20/2010] [Indexed: 11/17/2022] Open
Abstract
Dystroglycan is a central component of the dystrophin-glycoprotein complex (DGC) that links extracellular matrix with cytoskeleton, expressed in a variety of fetal and adult tissues. Dystroglycan plays diverse roles in development and homeostasis including basement membrane formation, epithelial morphogenesis, membrane stability, cell polarization, and cell migration. In this paper, we will focus on biological role of dystroglycan in Schwann cell function, especially myelination. First, we review the molecular architecture of DGC in Schwann cell abaxonal membrane. Then, we will review the loss-of-function studies using targeted mutagenesis, which have revealed biological functions of each component of DGC in Schwann cells. Based on these findings, roles of dystroglycan in Schwann cell function, in myelination in particular, and its implications in diseases will be discussed in detail. Finally, in view of the fact that understanding the role of dystroglycan in Schwann cells is just beginning, future perspectives will be discussed.
Collapse
|
31
|
Villagomez M, Szabo E, Podcheko A, Feng T, Papp S, Opas M. Calreticulin and focal-contact-dependent adhesion. Biochem Cell Biol 2009; 87:545-56. [PMID: 19767819 DOI: 10.1139/o09-016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell adhesion is regulated by a variety of Ca2+-regulated pathways that depend on Ca2+-binding proteins. One such protein is calreticulin, an ER-resident protein. Calreticulin signalling from within the ER can affect processes outside the ER, such as expression of several adhesion-related genes, most notably vinculin and fibronectin. In addition, changes in the expression level of calreticulin strongly affect tyrosine phosphorylation of cellular proteins, which is known to affect many adhesion-related functions. While calreticulin has been localized to cellular compartments other than the ER, it appears that only the ER-resident calreticulin affects focal-contact-dependent adhesion. In contrast, calreticulin residing outside the ER may be involved in contact disassembly and other adhesion phenomena. Here, we review the role of calreticulin in focal contact initiation, stabilization, and turnover. We propose that calreticulin may regulate cell-substratum adhesion by participating in an "ER-to-nucleus" signalling and in parallel "ER-to-cell surface" signalling based on posttranslational events.
Collapse
Affiliation(s)
- Maria Villagomez
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Toronto, ON M5S 1A8, Canada
| | | | | | | | | | | |
Collapse
|
32
|
Svendsen Ø, Barczyk M, Popova S, Lidén A, Gullberg D, Wiig H. The α11β1 Integrin Has a Mechanistic Role in Control of Interstitial Fluid Pressure and Edema Formation in Inflammation. Arterioscler Thromb Vasc Biol 2009; 29:1864-70. [DOI: 10.1161/atvbaha.109.194308] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Ø.S. Svendsen
- From the Department of Anesthesia and Intensive Care (Ø.S.S.), Haukeland University Hospital, and the Department of Biomedicine (Ø.S.S., M.M.B., S.N.P., Å.L., D.G., H.W.), University of Bergen, Norway
| | - M.M. Barczyk
- From the Department of Anesthesia and Intensive Care (Ø.S.S.), Haukeland University Hospital, and the Department of Biomedicine (Ø.S.S., M.M.B., S.N.P., Å.L., D.G., H.W.), University of Bergen, Norway
| | - S.N. Popova
- From the Department of Anesthesia and Intensive Care (Ø.S.S.), Haukeland University Hospital, and the Department of Biomedicine (Ø.S.S., M.M.B., S.N.P., Å.L., D.G., H.W.), University of Bergen, Norway
| | - Å Lidén
- From the Department of Anesthesia and Intensive Care (Ø.S.S.), Haukeland University Hospital, and the Department of Biomedicine (Ø.S.S., M.M.B., S.N.P., Å.L., D.G., H.W.), University of Bergen, Norway
| | - D. Gullberg
- From the Department of Anesthesia and Intensive Care (Ø.S.S.), Haukeland University Hospital, and the Department of Biomedicine (Ø.S.S., M.M.B., S.N.P., Å.L., D.G., H.W.), University of Bergen, Norway
| | - H. Wiig
- From the Department of Anesthesia and Intensive Care (Ø.S.S.), Haukeland University Hospital, and the Department of Biomedicine (Ø.S.S., M.M.B., S.N.P., Å.L., D.G., H.W.), University of Bergen, Norway
| |
Collapse
|
33
|
Studies on Multifunctional Effect of All-Trans Retinoic Acid (ATRA) on Matrix Metalloproteinase-2 (MMP-2) and Its Regulatory Molecules in Human Breast Cancer Cells (MCF-7). JOURNAL OF ONCOLOGY 2009; 2009:627840. [PMID: 19636436 PMCID: PMC2712868 DOI: 10.1155/2009/627840] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2008] [Revised: 02/28/2009] [Accepted: 05/01/2009] [Indexed: 11/17/2022]
Abstract
Background. Vitamin A derivative all-trans retinoic acid (ATRA) is considered as a potent chemotherapeutic drug for its capability of regulating cell growth and differentiation. We studied the effect of ATRA on MMP-2 in MCF-7, human breast cancer cells, and the probable signaling pathways which are affected by ATRA on regulating pro-MMP-2 activity and expression. Methods. Gelatin zymography, RT-PCR, ELISA, Western blot, Immunoprecipitation, and Cell adhesion assay are used. Results. Gelatin zymography showed that ATRA caused a dose-dependent inhibition of pro-MMP-2 activity. ATRA treatment downregulates the expression of MT1-MMP, EMMPRIN, FAK, NF-kB, and p-ERK. However, expression of E-cadherin, RAR, and CRABP increased upon ATRA treatment. Binding of cells to extra cellular matrix (ECM) protein fibronectin reduced significantly after ATRA treatment. Conclusions. The experimental findings clearly showed the inhibition of MMP-2 activity upon ATRA treatment. This inhibitory effect of ATRA on MMP-2 activity in human breast cancer cells (MCF-7) may result due to its inhibitory effect on MT1-MMP, EMMPRIN, and upregulation of TIMP-2. This study is focused on the effect of ATRA on MMP, MMP-integrin-E-cadherin interrelationship, and also the effect of the drug on different signaling molecules which may involve in the progression of malignant tumor development.
Collapse
|
34
|
Guignandon A, Akhouayri O, Usson Y, Rattner A, Laroche N, Lafage-Proust MH, Alexandre C, Vico L. Focal Contact Clustering in Osteoblastic Cells under Mechanical Stresses: Microgravity and Cyclic Deformation. ACTA ACUST UNITED AC 2009. [DOI: 10.1080/cac.10.2.69.83] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
35
|
Chen ZL, Haegeli V, Yu H, Strickland S. Cortical deficiency of laminin gamma1 impairs the AKT/GSK-3beta signaling pathway and leads to defects in neurite outgrowth and neuronal migration. Dev Biol 2009; 327:158-68. [PMID: 19118544 PMCID: PMC2669444 DOI: 10.1016/j.ydbio.2008.12.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2008] [Revised: 12/04/2008] [Accepted: 12/05/2008] [Indexed: 11/25/2022]
Abstract
Laminins have dramatic and varied actions on neurons in vitro. However, their in vivo function in brain development is not clear. Here we show that knockout of laminin gamma1 in the cerebral cortex leads to defects in neuritogenesis and neuronal migration. In the mutant mice, cortical layer structures were disrupted, and axonal pathfinding was impaired. During development, loss of laminin expression impaired phosphorylation of FAK and paxillin, indicating defects in integrin signaling pathways. Moreover, both phosphorylation and protein levels of GSK-3beta were significantly decreased, but only phosphorylation of AKT was affected in the mutant cortex. Knockout of laminin gamma1 expression in vitro, dramatically inhibited neurite growth. These results indicate that laminin regulates neurite growth and neuronal migration via integrin signaling through the AKT/GSK-3beta pathway, and thus reveal a novel mechanism of laminin function in brain development.
Collapse
Affiliation(s)
- Zu-Lin Chen
- Laboratory of Neurobiology and Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | | | | | | |
Collapse
|
36
|
Zhu J, Wang YS, Zhang J, Zhao W, Yang XM, Li X, Jiang TS, Yao LB. Focal adhesion kinase signaling pathway participates in the formation of choroidal neovascularization and regulates the proliferation and migration of choroidal microvascular endothelial cells by acting through HIF-1 and VEGF expression in RPE cells. Exp Eye Res 2008; 88:910-8. [PMID: 19111720 DOI: 10.1016/j.exer.2008.11.034] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2008] [Revised: 11/14/2008] [Accepted: 11/25/2008] [Indexed: 12/27/2022]
Abstract
Choroidal neovascularization (CNV) is one of the most frequent causes of severe and progressive vision loss, while its pathogenesis is still poorly understood. Focal adhesion kinase (FAK), a non-receptor tyrosine kinase, plays a crucial role in linking signals initiated by both the extracellular matrix (ECM) and soluble signaling factors and controls essential cellular processes. Extensive evidence has shown that FAK is activated in angiogenic response. This study aims to investigate the effect of FAK on CNV formation. The Brown-Norway (BN) rats underwent laser rupture of Bruch's membrane to induce CNV and were then killed at 1, 3, 7, and 14 days following laser injury. Immunofluorescence and Western blot were processed to detect FAK protein. Retinal pigment epithelial (RPE) cells were cultured under hypoxia and RNA interference (RNAi) technique was used to knock down the FAK gene in RPE cells. Expression of hypoxia inducible factor-1 (HIF-1alpha) and vascular endothelial growth factor (VEGF) in RPE cells were investigated by RT-PCR and Western blot. Two kinds of coculture models were used to observe the effects of specific blockade of FAK in RPE cells on the proliferation and migration of choroidal microvascular endothelial cells (CECs), respectively. FAK was highly expressed in the rat RPE-choroid tissue after photocoagulation. In vitro experiment showed that FAK was involved in hypoxia signaling in cultured RPE cells. The absence of FAK effectively reduced the expression of hypoxia-induced HIF-1alpha and VEGF in RPE cells, resulting in the inhibition of proliferation and migration of CECs. Our results suggest that FAK pathway activation plays a role in the development of CNV, and regulates the proliferation and migration of CECs by acting through HIF-1 and then up-regulating the expression of the angiogenic factor VEGF in RPE cells. It is reasonable to propose that FAK siRNA will potentially provides a means to attenuate the strong stimuli for neovascularization in CNV-dependent disorders, which could present a therapeutically relevant strategy for the inhibition of CNV.
Collapse
Affiliation(s)
- Jie Zhu
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Shaanxi Province, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Liu Z, Zhang HM, Yuan J, Lim T, Sall A, Taylor GA, Yang D. Focal adhesion kinase mediates the interferon-gamma-inducible GTPase-induced phosphatidylinositol 3-kinase/Akt survival pathway and further initiates a positive feedback loop of NF-kappaB activation. Cell Microbiol 2008; 10:1787-800. [PMID: 18452580 PMCID: PMC2765670 DOI: 10.1111/j.1462-5822.2008.01165.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Interferon-gamma-inducible GTPase (IGTP) expression is upregulated in coxsackievirus B3 (CVB3)-infected murine heart and inhibits CVB3-induced apoptosis through activation of the PI3 kinase/Akt pathway. However, the mechanism of this pathway activation is unknown. In this study, using doxcycycline-inducible Tet-On HeLa cells that overexpress IGTP, we have demonstrated that focal adhesion kinase (FAK) is phosphorylated in response to IGTP expression and that transfection of the Tet-On HeLa cells with a dominant negative FAK (FRNK) blocks Akt activation. Furthermore, induction of IGTP also promoted the NF-kappaB activation as evidenced by its enhanced nuclear translocation, binding to transcriptional promoters and increased transcriptional activity. However, FRNK transfection and phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 both blocked the IGTP-induced translocation and NF-kappaB activation. Furthermore, silencing NF-kappaB with siRNAs significantly inhibited the phosphorylation of FAK and Akt, but not their total expression levels, indicating that NF-kappaB activation is required for the IGTP-induced activation of FAK and PI3K/Akt. Finally, blocking this survival pathway by transfection of FRNK or silencing of NF-kappaB reduced CVB3 replication and enhanced cell death during CVB3 infection. Taken together, these results suggest that FAK is a mediator upstream of PI3K/Akt and NF-kappaB functions as a downstream effector and also positively regulates the activity of upstream kinases.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Pathology and Laboratory Medicine, University of British Columbia- The James Hogg iCAPTURE Center-St. Paul's Hospital, Vancouver, Canada
| | - Huifang M. Zhang
- Department of Pathology and Laboratory Medicine, University of British Columbia- The James Hogg iCAPTURE Center-St. Paul's Hospital, Vancouver, Canada
| | - Ji Yuan
- Department of Pathology and Laboratory Medicine, University of British Columbia- The James Hogg iCAPTURE Center-St. Paul's Hospital, Vancouver, Canada
| | - Travis Lim
- Department of Pathology and Laboratory Medicine, University of British Columbia- The James Hogg iCAPTURE Center-St. Paul's Hospital, Vancouver, Canada
| | - Alhousseynou Sall
- Department of Pathology and Laboratory Medicine, University of British Columbia- The James Hogg iCAPTURE Center-St. Paul's Hospital, Vancouver, Canada
| | - Gregory A. Taylor
- Geriatric Research, Education, and Clinical Center, VA Medical Center, Durham, North Carolina, USA
- Departments of Medicine and Immunology, Division of Geriatrics, and Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, North Carolina, USA
| | - Decheng Yang
- Department of Pathology and Laboratory Medicine, University of British Columbia- The James Hogg iCAPTURE Center-St. Paul's Hospital, Vancouver, Canada
| |
Collapse
|
38
|
Cardiac developmental defects and eccentric right ventricular hypertrophy in cardiomyocyte focal adhesion kinase (FAK) conditional knockout mice. Proc Natl Acad Sci U S A 2008; 105:6638-43. [PMID: 18448675 DOI: 10.1073/pnas.0802319105] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase that plays an important role in integrin-mediated signal transduction. To explore the role and mechanisms of FAK in cardiac development, we inactivated FAK in embryonic cardiomyocytes by crossing the floxed FAK mice with myosin light chain-2a (MLC2a) Cre mice, which expressed Cre as early as embryonic day 9.5 in the heart. The majority of conditional FAK knockout mice generated from MLC2a-Cre (CFKO-2a) died in the embryonic stage with thin ventricular wall and ventricular septal defects. A small fraction of CFKO-2a mice survived to adulthood with spontaneous eccentric right ventricle hypertrophy. Transmission electron microscopy analysis displayed swelling in the rough endoplasmic reticulum in CFKO-2a embryonic cardiomyocytes. We found that decreased cell proliferation, but not increased cell apoptosis or differentiation, is the reason for the thin ventricular wall in CFKO-2a mice. Microarray analysis suggests that myocyte enhancer factor 2a (MEF2a) can be regulated by FAK and that inactivation of FAK in the embryonic heart compromised MEF2a expression. Last, we found that Src, but not PI3K, is important in mediating signal transduction for the regulation of MEF2a by FAK. Together, these results identified the role and mechanisms of FAK in embryonic cardiac development.
Collapse
|
39
|
Liu P, Zhang C, Feng JB, Zhao YX, Wang XP, Yang JM, Zhang MX, Wang XL, Zhang Y. Cross talk among Smad, MAPK, and integrin signaling pathways enhances adventitial fibroblast functions activated by transforming growth factor-beta1 and inhibited by Gax. Arterioscler Thromb Vasc Biol 2008; 28:725-31. [PMID: 18187669 DOI: 10.1161/atvbaha.107.159889] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We investigated whether Smad, mitogen-activated protein kinase (MAPK), and integrin signaling pathways cross-talk to enhance adventitial fibroblast (AF) bioactivity, which was activated by transforming growth factor (TGF)-beta1 and inhibited by Gax. METHODS AND RESULTS Cultured AFs were stimulated with Ad-Gax, TGF-beta1, and siRNA-Gax. Assays for AFs viabilities demonstrated that TGF-beta1 and siRNA-Gax enhanced AFs proliferative, migratory, and adherent abilities, whereas Gax counteracted TGF-beta1-activated actions. Flow cytometry revealed that TGF-beta1 and siRNA-Gax increased S phase cells; however, Gax decreased AFs in the S phase and increased those in the G0-G1 and apoptotic phases. RT-PCR, Western blotting, and immunocytochemistry showed that TGF-beta1 and siRNA-Gax upregulated the expression of cytokines in Smad, MAPK, and integrin signaling pathways, and downregulated that of p15, p16, and p21. Conversely, Gax induced downregulation of these cytokines and upregulation of p15, p16, and p21. Thus, these signaling pathways cross-talk to enhance AF bioactivity; Gax effectively counteracts TGF-beta1 effects, blocks the cross-talk of these pathways, inhibits AF functions, and increases AF apoptosis. CONCLUSIONS Our findings indicate that cross-talk among Smad, MAPK, and integrin signaling pathways may account mainly for the mechanism of AF functions. Gax is a promising therapeutic gene for dissecting the signaling pathways controlling AF bioactivities.
Collapse
Affiliation(s)
- Ping Liu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Shandong University Qilu Hospital, Jinan, Shandong, China
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Grove M, Komiyama NH, Nave KA, Grant SG, Sherman DL, Brophy PJ. FAK is required for axonal sorting by Schwann cells. ACTA ACUST UNITED AC 2007; 176:277-82. [PMID: 17242067 PMCID: PMC2063954 DOI: 10.1083/jcb.200609021] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Signaling by laminins and axonal neuregulin has been implicated in regulating axon sorting by myelin-forming Schwann cells. However, the signal transduction mechanisms are unknown. Focal adhesion kinase (FAK) has been linked to α6β1 integrin and ErbB receptor signaling, and we show that myelination by Schwann cells lacking FAK is severely impaired. Mutant Schwann cells could interdigitate between axon bundles, indicating that FAK signaling was not required for process extension. However, Schwann cell FAK was required to stimulate cell proliferation, suggesting that amyelination was caused by insufficient Schwann cells. ErbB2 receptor and AKT were robustly phosphorylated in mutant Schwann cells, indicating that neuregulin signaling from axons was unimpaired. These findings demonstrate the vital relationship between axon defasciculation and Schwann cell number and show the importance of FAK in regulating cell proliferation in the developing nervous system.
Collapse
Affiliation(s)
- Matthew Grove
- Centre for Neuroscience Research, University of Edinburgh, Edinburgh EH9 1QH, Scotland, UK
| | | | | | | | | | | |
Collapse
|
41
|
Chen H, Bai J, Ye J, Liu Z, Chen R, Mao W, Li A, Zhou J. JWA as a functional molecule to regulate cancer cells migration via MAPK cascades and F-actin cytoskeleton. Cell Signal 2007; 19:1315-27. [PMID: 17336041 DOI: 10.1016/j.cellsig.2007.01.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2006] [Revised: 01/07/2007] [Accepted: 01/07/2007] [Indexed: 11/21/2022]
Abstract
Mitogen activated protein kinase (MAPK) cascades are thought to mediate diverse biological functions such as cell growth, differentiation and migration. Activated MAPK may affect microtubule (MT) which is essential for cellular polarity, differentiation and motility. Data in this study show that JWA, a newly identified novel microtubule-associated protein (MAP) was essential for the rearrangement of F-actin cytoskeleton and activation of MAPK cascades induced by arsenic trioxide (As2O3) and phorbol ester (PMA). Over-expression of JWA alone in HeLa, B16 and HCCLM3 cancer cells effectively inhibited cellular migration; whereas, cellular migration was significantly accelerated when cells were deficient in JWA expression. The mechanism underlying these phenomena might be due to JWA affected F-actin rearrangement. Furthermore, JWA deficiency blocked anti-migratory effect produced by As2O3 but enhanced the migratory effect initiated by PMA in HeLa cells. JWA SDR-SLR motifs are not only critical for the MAPK cascades activation, but also for cell migration. Further studies found that JWA differentially regulated cell migration via ERK downstream effectors focal adhesion kinase (FAK) and cyclooxygenase-2 (COX-2). Therefore, JWA regulated-tumor cellular migration might involve MAPK cascades activation and F-actin cytoskeleton rearrangement mechanisms. Our data provide an unexpected role for JWA in tumor cell migration behaviors.
Collapse
Affiliation(s)
- Hairong Chen
- Department of Molecular Cell Biology and Toxicology, School of Public Health, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Rice KM, Desai DH, Kinnard RS, Harris R, Wright GL, Blough ER. Load-induced focal adhesion mechanotransduction is altered with aging in the Fischer 344/NNiaHSd x Brown Norway/BiNia rat aorta. Biogerontology 2006; 8:257-67. [PMID: 17136425 DOI: 10.1007/s10522-006-9066-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2006] [Accepted: 10/02/2006] [Indexed: 11/29/2022]
Abstract
The focal adhesion kinase (FAK) pathway has emerged as a critical component for mediating numerous cellular responses including control of cell growth, differentiation, and adaptation. Here we compared the expression, basal activation, and the ability of increased intraluminal pressure to activate FAK and focal adhesion-associated proteins in the aorta of adult (6 months old) and very aged (36 months old) Fischer 344/NNiaHSd x Brown Norway/BiNia (F344/NXBN) rats. Immunoblot analysis showed increases in the aortic content of FAK (15%), FAK related non-kinase (p41-FRNK) (28%), Src (92%), RhoA (41%), and paxillin (23%) in the very aged aortae. Increased age significantly changed the basal phosphorylation status of FAK and paxillin. Application of aortic intraluminal pressure (200 mm Hg) amplified the phosphorylation of FAK (Tyr 925), Src (Tyr 416), and paxillin (Tyr 188) in adult animals while aortic loading in the very aged animals failed to induce FAK (Tyr 925) phosphorylation. Aging did not alter the load-induced regulation of RhoA; however, FRNK (p41) translocation between cytosolic and membrane compartments was increased. These results confirm previous observations that FAK and focal adhesion-associated proteins are mechanically regulated and expand these studies to suggest that FAK mechanotransduction is altered with aging.
Collapse
Affiliation(s)
- K M Rice
- Department of Pharmacology, Physiology, and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755-1090, USA
| | | | | | | | | | | |
Collapse
|
43
|
Jiang X, Sinnett-Smith J, Rozengurt E. Differential FAK phosphorylation at Ser-910, Ser-843 and Tyr-397 induced by angiotensin II, LPA and EGF in intestinal epithelial cells. Cell Signal 2006; 19:1000-10. [PMID: 17240116 PMCID: PMC1868572 DOI: 10.1016/j.cellsig.2006.11.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2006] [Revised: 11/13/2006] [Accepted: 11/16/2006] [Indexed: 12/20/2022]
Abstract
A rapid increase in the tyrosine phosphorylation of the non-receptor tyrosine kinase FAK is a prominent early event in fibroblasts stimulated by a variety of signaling molecules. However, a variety of epithelial cells, including intestinal epithelial cells, show a high basal level of tyrosine phosphorylated FAK that is only slightly further increased by addition of G protein-coupled receptor (GPCR) agonists or growth factors. In this study, we determined whether these stimuli could elicit FAK phosphorylation at serine residues, including Ser-910 and Ser-843. Our results show that multiple agonists including angiotensin II (ANGII), lysophosphatidic acid (LPA), phorbol esters and EGF induced a striking stimulation of FAK phosphorylation at Ser-910 in rat intestinal epithelial IEC-18 cells via an ERK-dependent pathway. In striking contrast, none of these stimuli promoted a significant further increase in FAK phosphorylation at Tyr-397 in these cells. These results were extended using cultures of polarized human colonic epithelial T84 cells. We found that either carbachol or EGF promoted a striking ERK-dependent phosphorylation of FAK at Ser-910, but these agonists caused only slight stimulation of FAK at Tyr-397 in T84 cells. In addition, we demonstrated that GPCR agonists also induced a dramatic increase of FAK phosphorylation at Ser-843 in either IEC-18 or T84 cells. Our results indicate that Ser-910 and Ser-843, rather than Tyr-397, are prominent sites differentially phosphorylated in response to neurotransmitters, bioactive lipids, tumor promoters and growth factors in intestinal epithelial cells.
Collapse
Affiliation(s)
- Xiaohua Jiang
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, CURE: Digestive Diseases Research Center and Molecular Biology Institute, University of California, Los Angeles, California 90095-178622, USA
| | | | | |
Collapse
|
44
|
Miłoszewska J, Szaniawska B, Trembacz H, Gos M, Przybyszewska M, Swoboda P, Małecki M, Janik P. Effect of transfection with a gene coding for the fibronectin FNIII/10 fragment upon contact inhibition of C3H10T1/2 fibroblasts. Cell Prolif 2006; 39:195-203. [PMID: 16671997 PMCID: PMC6496633 DOI: 10.1111/j.1365-2184.2006.00382.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The density-dependent growth inhibition of non-transformed cells may be associated with inefficient transduction of the proliferative signal from cell adhesion molecules. To verify this concept, the C3H10T1/2 fibroblasts were stably transfected with the gene coding for the fibronectin fragment III/10 (FNIII/10). This resulted in differences in gene's expression between original C3H10T1/2 cells and their FNIII/10 transfectants. No significant differences in growth properties were observed in the original or in the transfected cells. C3H10T1/2 cells and their transfectants, when co-cultured, displayed more cells at confluence than the cells cultured alone. Moreover, co-cultured C3H10T1/2 cells and their transfectants showed elevated levels of phospho-ERK1/2 compared to homogenous cultures. Results obtained indicate that cellular homogeneity is responsible for density-dependent growth inhibition.
Collapse
Affiliation(s)
- J Miłoszewska
- Cell Biology Department, Cancer Center, W.K. Roentgen 5, 02-781 Warsaw, Poland
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Jiang X, Jacamo R, Zhukova E, Sinnett-Smith J, Rozengurt E. RNA interference reveals a differential role of FAK and Pyk2 in cell migration, leading edge formation and increase in focal adhesions induced by LPA in intestinal epithelial cells. J Cell Physiol 2006; 207:816-28. [PMID: 16508947 DOI: 10.1002/jcp.20629] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
In the gastrointestinal mucosa, cell migration plays a crucial role in the organization and maintenance of tissue integrity but the mechanisms involved remain incompletely understood. Here, we used small-interfering RNA (siRNA)-mediated depletion of focal adhesion kinase (FAK) protein to determine the role of FAK in wound-induced migration and cytoskeletal organization in the non-transformed intestinal epithelial cells IEC-6 and IEC-18 stimulated with the G protein-coupled receptors (GPCR) agonist lysophosphatidic acid (LPA). Treatment of these cells with FAK siRNA substantially reduced FAK expression, but did not affect the expression of proline-rich tyrosine kinase 2 (Pyk2). Knockdown of FAK protein significantly inhibited LPA-induced migration of both IEC-18 and IEC-6 cells. LPA induced reorganization of actin and microtubule cytoskeleton in the leading edge was largely inhibited in FAK siRNA-transfected IEC-18 cells. Interestingly, in contrast to the FAK-/- cells, which exhibit an increased number of prominent focal adhesions when plated on fibronectin, FAK knockdown IEC-18 cells exhibited dramatically decreased number of focal adhesions in response to both LPA and fibronectin as compared with the control cells. We also used siRNAs to knockdown Pyk2 expression without reducing FAK expression. Depletion of Pyk2 did not prevent LPA-induced migration or cytoskeletal reorganization in IEC-18 cells. In conclusion, our study shows that FAK plays a critical role in LPA-induced migration, cytoskeletal reorganization, and assembly of focal adhesions in intestinal epithelial cells whereas depletion of Pyk2 did not interfere with any of these responses elicited by LPA.
Collapse
Affiliation(s)
- Xiaohua Jiang
- Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, CURE: Digestive Diseases Research Center and Molecular Biology Institute, University of California, Los Angeles, California 90095-178622, USA
| | | | | | | | | |
Collapse
|
46
|
Saunders RM, Holt MR, Jennings L, Sutton DH, Barsukov IL, Bobkov A, Liddington RC, Adamson EA, Dunn GA, Critchley DR. Role of vinculin in regulating focal adhesion turnover. Eur J Cell Biol 2006; 85:487-500. [PMID: 16584805 DOI: 10.1016/j.ejcb.2006.01.014] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2005] [Revised: 12/30/2005] [Accepted: 01/02/2006] [Indexed: 11/15/2022] Open
Abstract
Although vinculin (-/-) mouse embryo fibroblasts assemble focal adhesions (FAs), they spread more slowly, less extensively, and close a wound more rapidly than vinculin (+/+) cells. To investigate the structure and dynamics of FAs in these cells, we used real-time interference reflection microscopy (IRM) thus avoiding the need to express exogenous GFP-tagged FA proteins which may be misregulated. This showed that the FAs were smaller, less abundant and turned over more rapidly in vinculin null compared to wild-type cells. Expression of vinculin rescued the spreading defect and resulted in larger and more stable FAs. Phosphatidylinositol 4,5-bisphosphate (PIP2) is thought to play a role in vinculin activation by relieving an intramolecular association between the vinculin head (Vh) and tail (Vt) that masks the ligand binding sites in Vh and Vt. To investigate the role of the vinculin/PIP2 interaction in FA dynamics, we used a vinculin mutant lacking the C-terminal arm (residues 1053-1066) and referred to as the deltaC mutation. This mutation reduced PIP2 binding to a Vt deltaC polypeptide by >90% compared to wild type without affecting binding to Vh or F-actin. Interestingly, cells expressing the vinculin deltaC mutant assembled remarkably stable FAs. The results suggest that vinculin inhibits cell migration by stabilising FAs, and that binding of inositol phospholipids to Vt plays an important role in FA turnover.
Collapse
Affiliation(s)
- Ruth M Saunders
- Department of Biochemistry, University of Leicester, University Road, Leicester LE1 7RH, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Bouchard MJ, Wang L, Schneider RJ. Activation of focal adhesion kinase by hepatitis B virus HBx protein: multiple functions in viral replication. J Virol 2006; 80:4406-14. [PMID: 16611900 PMCID: PMC1472019 DOI: 10.1128/jvi.80.9.4406-4414.2006] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The hepatitis B virus (HBV) X protein (HBx) is a multifunctional regulator of cellular signal transduction and transcription pathways and has a critical role in HBV replication. Much of the cytoplasmic signal transduction activity associated with HBx expression and its stimulation of viral replication is attributable to HBx-induced activation of calcium signaling pathways involving Pyk2 and Src tyrosine kinases. To further characterize upstream signal transduction pathways that are required for HBx activity, including activation of Src and mitogen-activated protein kinase (MAPK) cascades, we determined whether focal adhesion kinase (FAK), a known regulator of Src family kinases and the other member of the Pyk2/FAK kinase family, is activated by HBx. We report that HBx activates FAK and that FAK activation is important for multiple HBx functions. Dominant inhibiting forms of FAK blocked HBx activation of Src kinases and downstream signal transduction, HBx stimulation of NF-kappaB and AP-1-dependent transcription, and HBV DNA replication. We also demonstrate that HBx-induced activation of FAK is dependent on cellular calcium signaling, which is modulated by HBx. Moreover, prolonged expression of HBx increases both FAK activity and its level of expression. FAK activation may play a role in cellular transformation and cancer progression. HBx stimulation of FAK activity and abundance may also be relevant as a potential cofactor in HBV-associated hepatocellular carcinoma.
Collapse
Affiliation(s)
- Michael J Bouchard
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, USA
| | | | | |
Collapse
|
48
|
Szabo E, Papp S, Opas M. Calreticulin and cellular adhesion/migration-specific signalling pathways. J Appl Biomed 2006. [DOI: 10.32725/jab.2006.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
49
|
Mueller C, Emmrich J, Jaster R, Braun D, Liebe S, Sparmann G. Cis-hydroxyproline-induced inhibition of pancreatic cancer cell growth is mediated by endoplasmic reticulum stress. World J Gastroenterol 2006; 12:1569-76. [PMID: 16570349 PMCID: PMC4124289 DOI: 10.3748/wjg.v12.i10.1569] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the biological effects of cis-hydroxyproline (CHP) on the rat pancreatic carcinoma cell line DSL6A, and to examine the underlying molecular mechanisms.
METHODS: The effect of CHP on DSL6A cell proliferation was assessed by using BrdU incorporation. The expression of focal adhesion kinase (FAK) was characterized by Western blotting and immunofluorescence. Induction of endoplasmic reticulum (ER) stress was investigated by using RT-PCR and Western blotting for the glucose-related protein-78 (GRP78) and growth arrest and DNA inducible gene (GADD153). Cell viability was determined through measuring the metabolic activity based on the reduction potential of DSL6A cells. Apoptosis was analyzed by detection of caspase-3 activation and cleavage of poly(ADP-ribose) polymerase (PARP) as well as DNA laddering.
RESULTS: In addition to inhibition of proliferation, incubation with CHP induced proteolytic cleavage of FAK and a delocalisation of the enzyme from focal adhesions, followed by a loss of cell adherence. Simultaneously, we could show an increased expression of GRP78 and GADD153, indicating a CHP-mediated activation of the ER stress cascade in the DSL6A cell line. Prolonged incubation of DSL6A cells with CHP finally resulted in apoptotic cell death. Beside L-proline, the inhibition of intracellular proteolysis by addition of a broad spectrum protease inhibitor could abolish the effects of CHP on cellular functions and the molecular processes. In contrast, impeding the activity of apoptosis-executing caspases had no influence on CHP-mediated cell damage.
CONCLUSION: Our data suggest that the initiation of ER stress machinery by CHP leads to an activation of intracellular proteolytic processes, including caspase-independent FAK degradation, resulting in damaging pancreatic carcinoma cells.
Collapse
Affiliation(s)
- Christoph Mueller
- Department of Medicine, Division of Gastroenterology, University of Rostock, Ernst-Heydemann-Str. 6, D-18057 Rostock, Germany
| | | | | | | | | | | |
Collapse
|
50
|
Jacamo R, Jiang X, Lunn JA, Rozengurt E. FAK phosphorylation at Ser-843 inhibits Tyr-397 phosphorylation, cell spreading and migration. J Cell Physiol 2006; 210:436-44. [PMID: 17096371 DOI: 10.1002/jcp.20870] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Multiple stimuli promote the tyrosine phosphorylation and activation of focal adhesion kinase (FAK), which ultimately facilitates migration. Little is known about the effect of adhesion-dependent signals and cytoskeleton organization on the regulation of FAK phosphorylation at serine sites, or about the role of FAK serine phosphorylation in cell migration. Here, we show that FAK phosphorylation at Ser-843 is strikingly increased when adherent cells are removed from the substratum and held in suspension or by treatment of adherent cells with cytochalasin D, conditions that disrupt the F-actin cytoskeleton and promote focal adhesion disassembly. Notably, the increase in Ser-843 phosphorylation was accompanied by a concomitant sharp decrease in Tyr-397 phosphorylation. To further examine the cause-effect relationship between these two phosphorylation sites we generated Ser-843 phosphorylation-deficient and phosphorylation-mimicking FAK mutants. We found that mutation of Ser-843 to aspartic acid (FAK[S843D]) markedly decreased FAK Tyr-397 phosphorylation in integrin-stimulated cells. While the migratory defect of FAK-deficient fibroblasts was rescued by stable re-expression of WT FAK or FAK[S843A], stable re-expression of FAK[S843D] failed to restore the ability of the cells to migrate into the denuded area of a wound. Our results indicate that increased FAK phosphorylation at Ser-843 represses FAK phosphorylation at Tyr-397, thus suggesting a mechanism of cross-talk between these phosphorylation sites that could regulate FAK-mediated cell shape and migration.
Collapse
Affiliation(s)
- Rodrigo Jacamo
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, CURE: Digestive Diseases Research Center and Molecular Biology Institute, University of California, Los Angeles, California 90095, USA
| | | | | | | |
Collapse
|