1
|
Matsushige C, Kitazumi K, Beaman A, Miyagi M, Tallquist MD, Yamazaki Y. RGD peptide promotes follicle growth through integrins αvβ3/αvβ5 in three-dimensional culture. Reproduction 2025; 169:e240151. [PMID: 39441765 PMCID: PMC11729052 DOI: 10.1530/rep-24-0151] [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: 05/08/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 10/25/2024]
Abstract
In brief Three-dimensional ovarian tissue culture is a unique model to define the effects of molecules on folliculogenesis. Using this model, we determined that RGD-integrin interaction plays a role in antrum formation and theca cell differentiation. Abstract We recently developed a three-dimensional (3D) ovarian tissue culture system supported by bacterial-derived dextran hydrogel. Arg-Gly-Asp (RGD) is an extracellular matrix-derived triple peptide. Immature ovarian tissues cultured in RGD-modified dextran hydrogel significantly promoted antral follicle growth and oocyte quality compared with those cultured in dextran hydrogel alone. In this study, we examined the mechanism of follicle growth stimulated by RGD treatment in the 3D system. First, we detected that direct contact between RGD-modified dextran hydrogel and ovarian interstitial cells is necessary to promote antral follicle growth. Therefore, we hypothesized that RGD stimulates antral follicle growth through RGD-binding integrin receptors expressed in the interstitial cell mass. Using quantitative PCR (qPCR) and immunochemical staining, we identified that integrins ⍺vβ3 and ⍺v5 are predominantly expressed in the ovarian interstitial compartment. To assess the effect of RGD-integrin interaction on follicle growth, ovarian tissues were cultured with cilengitide (Ci), an inhibitor specific for ⍺vβ3 and ⍺vβ5. Ci treatment suppressed RGD-induced follicle growth and oocyte quality in a dose-dependent manner. When the interstitial cell aggregates were cultured with RGD, cell migration and theca-related gene expression were significantly upregulated. Ci treatment dramatically suppressed these RGD-induced activities. In coculturing the interstitial aggregate and secondary follicles with RGD, migrating cells formed the outermost cell layers around the follicles, like theca layers, which were totally blocked by Ci treatment. In conclusion, our results suggest that RGD stimulates theca cell differentiation in the ovarian interstitial cells through integrins ⍺vβ3 and ⍺v5 to promote antral follicle growth in our 3D system.
Collapse
Affiliation(s)
- Cassandra Matsushige
- Yanagimachi Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Kaelyn Kitazumi
- Yanagimachi Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Amanda Beaman
- Yanagimachi Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Marissa Miyagi
- Yanagimachi Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Michelle D Tallquist
- Center for Cardiovascular Research, Department of Medicine, John A Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Yukiko Yamazaki
- Yanagimachi Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| |
Collapse
|
2
|
Cappelleri A, Brambilla E, Chiti LE, Trapletti A, Bianchi GBM, Di Giancamillo M, Grieco V, Giudice C. Expression of α v Integrin in Feline Injection-Site Sarcoma (FISS): Preliminary Investigations. Animals (Basel) 2024; 14:3588. [PMID: 39765492 PMCID: PMC11672646 DOI: 10.3390/ani14243588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/27/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Feline injection-site sarcomas (FISSs) are malignant skin tumors of mesenchymal origin arising at local post-vaccination (or injection) sites. In recent years, a fluorescence imaging technique based on probes targeting αvβ3 integrin has been effectively applied for the surgical complete resection of the tumor. In our study, we investigated the utility of a commercially available anti-αv integrin polyclonal antibody for the histopathological evaluation of FISS's surgical excision margins. We collected 10 formalin-fixed paraffin-embedded (FFPE) feline excisional biopsies with a histopathological diagnosis of FISS (7 fibrosarcomas and 3 pleomorphic sarcomas) and wide margin tissue, along with one subcutaneous injection-site granuloma and 6 osteosarcomas. Samples were processed for histology, and slides were stained for IHC with the anti-αv integrin antibody. Immunostained slides were evaluated for the cellular localization and intensity of the staining in different neoplastic and non-neoplastic cell populations. Neoplastic and non-neoplastic spindle cells had cytoplasmic positivity in all fibrosarcomas. Inflammatory cells, including macrophages of the injection-site granuloma, were negative. Multinucleated giant cells in the pleomorphic sarcomas had an intense membranous positivity. Although the anti-αv integrin antibody was ineffective for the histopathological evaluation of surgical excision margins, the membranous localization of αv integrin in multinucleated giant cells of pleomorphic sarcomas suggests that it plays a role in the oncogenesis of this FISS variant.
Collapse
Affiliation(s)
- Andrea Cappelleri
- Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900 Lodi, Italy; (A.C.); (E.B.); (A.T.); (G.B.M.B.); (M.D.G.); (V.G.)
| | - Eleonora Brambilla
- Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900 Lodi, Italy; (A.C.); (E.B.); (A.T.); (G.B.M.B.); (M.D.G.); (V.G.)
| | - Lavinia E. Chiti
- Clinic for Small Animal Surgery, Department for Small Animals, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland;
| | - Alessia Trapletti
- Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900 Lodi, Italy; (A.C.); (E.B.); (A.T.); (G.B.M.B.); (M.D.G.); (V.G.)
| | - Gaia B. M. Bianchi
- Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900 Lodi, Italy; (A.C.); (E.B.); (A.T.); (G.B.M.B.); (M.D.G.); (V.G.)
| | - Mauro Di Giancamillo
- Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900 Lodi, Italy; (A.C.); (E.B.); (A.T.); (G.B.M.B.); (M.D.G.); (V.G.)
| | - Valeria Grieco
- Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900 Lodi, Italy; (A.C.); (E.B.); (A.T.); (G.B.M.B.); (M.D.G.); (V.G.)
| | - Chiara Giudice
- Department of Veterinary Medicine and Animal Sciences, University of Milan, 26900 Lodi, Italy; (A.C.); (E.B.); (A.T.); (G.B.M.B.); (M.D.G.); (V.G.)
| |
Collapse
|
3
|
Cui Y, Rolova T, Fagerholm SC. The role of integrins in brain health and neurodegenerative diseases. Eur J Cell Biol 2024; 103:151441. [PMID: 39002282 DOI: 10.1016/j.ejcb.2024.151441] [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: 04/29/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/15/2024] Open
Abstract
Integrins are heterodimeric membrane proteins expressed on the surface of most cells. They mediate adhesion and signaling processes relevant for a wealth of physiological processes, including nervous system development and function. Interestingly, integrins are also recognized therapeutic targets for inflammatory diseases, such as multiple sclerosis. Here, we discuss the role of integrins in brain development and function, as well as in neurodegenerative diseases affecting the brain (Alzheimer's disease, multiple sclerosis, stroke). Furthermore, we discuss therapeutic targeting of these adhesion receptors in inflammatory diseases of the brain.
Collapse
Affiliation(s)
- Yunhao Cui
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland
| | - Taisia Rolova
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki 00290, Finland
| | - Susanna C Fagerholm
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland.
| |
Collapse
|
4
|
Gondhane A, Verma P, Chandak A, Basu S. Prospective evaluation of 68 Ga-NODAGA-RGD PET-CT in patients of carcinoma thyroid with thyroglobulin elevated negative radioiodine scintigraphy (TENIS) with a head-to-head comparison with FDG-PET/CT. Nucl Med Commun 2024; 45:412-419. [PMID: 38445350 DOI: 10.1097/mnm.0000000000001828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
BACKGROUND AND AIM This study aimed to examine the expression of RGD binding integrins in patients of elevated serum thyroglobulin (Tg) level with negative radioiodine scintigraphy (TENIS) employing 68 Ga-NODAGA-RGD PET-CT. MATERIAL AND METHODS This was a prospective study involving 30 proven cases of TENIS with histopathological diagnosis of differentiated thyroid carcinoma post-surgery. In addition to observing the lesional concentration on 68 Ga-NODAGA-RGD PET-CT, a 4-point visual grading system (grade I-IV), was undertaken to estimate the degree of radiotracer avidity, for potential of theranostics. RESULTS On 18 F-FDG-PET/CT, the uptake was seen in 182 lesions out of a total of 200 (91%). 68 Ga-NODAGA-RGD PET-CT showed expression in a total of 110/200 (55%) lesions. On patient-specific analysis, 68 Ga-NODAGA-RGD PET-CT was positive for the disease in 21/30 patients (70%) and negative in 9/30 (30%) patients. The overall patient-specific sensitivity and specificity of 68 Ga-NODAGA-RGDPET-CT were 75% and 100%, respectively. 18 F-FDG PET-CT was positive for the disease in 26/30 patients (86.66%) and negative in 4/30 (13.33%) patients. The overall patient-specific sensitivity and specificity of 18 F-FDG-PET/CT were 92.86% and 100%, respectively. The 4-point visual grading system revealed 14/200 (7%) lesions demonstrating Grade I uptake, 49/200 (24.5%) lesions grade II uptake, 17/200 (8.5%) lesions grade III uptake and 40/200 (20%) lesions grade IV uptake. CONCLUSION The results suggested that RGD-binding integrin is expressed in a sizeable fraction of metastatic lesions of TENIS cases, albeit demonstrating a varying degree of uptake. Out of the soft tissue, lung, and bone lesions, metastatic bone lesions showed more RGD affinity than other sites. The patients with substantial RGD uptake on a 4-point visual grading system may be potential targets for RGD-based therapy.
Collapse
Affiliation(s)
- Abhay Gondhane
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, Parel and
- Homi Bhabha National Institute, Mumbai, India
| | - Priyanka Verma
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, Parel and
- Homi Bhabha National Institute, Mumbai, India
| | - Ashok Chandak
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, Parel and
| | - Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, Parel and
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
5
|
Ju Y, Shen T, Guo Z, Kong Y, Huang Y, Hu J. Vitronectin promotes insulin resistance in trophoblast cells by activating JNK in gestational diabetes mellitus. Cell Biol Int 2024. [PMID: 38654431 DOI: 10.1002/cbin.12167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 03/13/2024] [Accepted: 04/12/2024] [Indexed: 04/26/2024]
Abstract
Gestational diabetes mellitus (GDM) is a common disorder in the clinic, which may lead to severe detrimental outcomes both for mothers and infants. However, the underlying mechanisms for GDM are still not clear. In the present study, we performed label-free proteomics using placentas from GDM patients and normal controls. Vitronectin caused our attention among differentially expressed proteins due to its potential role in the pathological progression of GDM. Vitronectin was increased in the placentas of GDM patients, which was confirmed by Western blot analysis. Vitronectin represses insulin signal transduction in trophoblast cells, whereas the knockdown of vitronectin further potentiates insulin-evoked events. Neutralization of CD51/61 abolishes the repressed insulin signal transduction in vitronectin-treated trophoblast cells. Moreover, vitronectin activates JNK in a CD51/61-depedent manner. Inhibition of JNK rescues impaired insulin signal transduction induced by vitronectin. Overall, our data indicate that vitronectin binds CD51/61 in trophoblast cells to activate JNK, and thus induces insulin resistance. In this regard, increased expression of vitronectin is likely a risk factor for the pathological progression of GDM. Moreover, blockade of vitronectin production or its receptors (CD51/61) may have therapeutic potential for dealing with GDM.
Collapse
Affiliation(s)
- Yuejun Ju
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
- Department of Endocrinology, Changshu No.2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu, P.R. China
| | - Ting Shen
- Department of Endocrinology, Changshu No.2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu, P.R. China
| | - Zhanhong Guo
- Department of Endocrinology, Changshu No.2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu, P.R. China
| | - Yinghong Kong
- Department of Endocrinology, Changshu No.2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Jiangsu, P.R. China
| | - Yun Huang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Ji Hu
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| |
Collapse
|
6
|
Wang Y, Schneider SW, Gorzelanny C. Crosstalk between Circulating Tumor Cells and Plasma Proteins-Impact on Coagulation and Anticoagulation. Cancers (Basel) 2023; 15:cancers15113025. [PMID: 37296987 DOI: 10.3390/cancers15113025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/24/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Cancer metastasis is a complex process. After their intravasation into the circulation, the cancer cells are exposed to a harsh environment of physical and biochemical hazards. Whether circulating tumor cells (CTCs) survive and escape from blood flow defines their ability to metastasize. CTCs sense their environment with surface-exposed receptors. The recognition of corresponding ligands, e.g., fibrinogen, by integrins can induce intracellular signaling processes driving CTCs' survival. Other receptors, such as tissue factor (TF), enable CTCs to induce coagulation. Cancer-associated thrombosis (CAT) is adversely connected to patients' outcome. However, cancer cells have also the ability to inhibit coagulation, e.g., through expressing thrombomodulin (TM) or heparan sulfate (HS), an activator of antithrombin (AT). To that extent, individual CTCs can interact with plasma proteins, and whether these interactions are connected to metastasis or clinical symptoms such as CAT is largely unknown. In the present review, we discuss the biological and clinical relevance of cancer-cell-expressed surface molecules and their interaction with plasma proteins. We aim to encourage future research to expand our knowledge of the CTC interactome, as this may not only yield new molecular markers improving liquid-biopsy-based diagnostics but also additional targets for better cancer therapies.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Stefan W Schneider
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Christian Gorzelanny
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| |
Collapse
|
7
|
Cossu J, Thoreau F, Boturyn D. Multimeric RGD-Based Strategies for Selective Drug Delivery to Tumor Tissues. Pharmaceutics 2023; 15:pharmaceutics15020525. [PMID: 36839846 PMCID: PMC9961187 DOI: 10.3390/pharmaceutics15020525] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
RGD peptides have received a lot of attention over the two last decades, in particular to improve tumor therapy through the targeting of the αVβ3 integrin receptor. This review focuses on the molecular design of multimeric RGD compounds, as well as the design of suitable linkers for drug delivery. Many examples of RGD-drug conjugates have been developed, and we show the importance of RGD constructs to enhance binding affinity to tumor cells, as well as their drug uptake. Further, we also highlight the use of RGD peptides as theranostic systems, promising tools offering dual modality, such as tumor diagnosis and therapy. In conclusion, we address the challenging issues, as well as ongoing and future development, in comparison with large molecules, such as monoclonal antibodies.
Collapse
Affiliation(s)
- Jordan Cossu
- University Grenoble Alpes, CNRS, DCM UMR 5250, F-38000 Grenoble, France
| | - Fabien Thoreau
- University Poitiers, Inst Chim Milieux & Mat Poitiers IC2MP, UMR CNRS 7285, F-86073 Poitiers, France
| | - Didier Boturyn
- University Grenoble Alpes, CNRS, DCM UMR 5250, F-38000 Grenoble, France
- Correspondence:
| |
Collapse
|
8
|
Matsushige C, Xu X, Miyagi M, Zuo YY, Yamazaki Y. RGD-modified dextran hydrogel promotes follicle growth in three-dimensional ovarian tissue culture in mice. Theriogenology 2022; 183:120-131. [PMID: 35247849 PMCID: PMC9005264 DOI: 10.1016/j.theriogenology.2022.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 10/19/2022]
Abstract
In vitro follicle growth is a promising technology to preserve fertility for cancer patients. We previously developed a three-dimensional (3-D) ovarian tissue culture system supported by mouse tumor cell-derived Matrigel. When murine ovarian tissues at 14 days old were cultured in Matrigel drops, antrum formation and oocyte competence were significantly enhanced compared with those cultured without Matrigel. In this study, we tested whether nonanimal-derived dextran hydrogels can support a 3-D ovarian tissue culture. We employed chemically defined dextran hydrogels consisting of dextran polymers crosslinked with polyethylene glycol (PEG)-based cell-degradable crosslinker. To determine the optimal gel elasticity for the 3-D tissue culture, we measured Young's modulus of dextran hydrogels at four concentrations (1.75, 2.25, 2.75, and 3.25 mmol/L), and cultured ovarian tissues in these gels for 7 days. As a result, 2.25 mmol/L dextran hydrogel with Young's modulus of 224 Pa was appropriate to provide physical support as well as to promote follicle expansion in the 3-D system. To mimic the natural extracellular matrix (ECM) environment, we modified the dextran hydrogels with two bioactive factors: ECM-derived Arg-Gly-Asp (RGD) peptides as a cell-adhesive factor, and activin A. The ovarian tissues were cultured in 2.25 mmol/L dextran hydrogels under four different conditions: Activin-/RGD- (A-R-), A + R-, A-R+, and A + R+. On Day 7 of culture, follicle and oocyte sizes were significantly increased in the RGD-modified conditions compared with those without RGD. The RGD-modified hydrogels also promoted mRNA levels of steroidogenic-related genes and estradiol production in the 3-D ovarian tissue culture. In vitro maturation and developmental competence of follicular oocytes were remarkably improved in the presence of RGD. In particular, blastocyst embryos were obtained only from A-R+ or A+R+ conditions after in vitro fertilization. We also determined synergistic effects of the RGD peptides and activin A on follicle growth and oocyte development in the 3-D tissue culture. In conclusion, our results suggest that RGD-modified dextran hydrogels provide an ECM-mimetic bioactive environment to support folliculogenesis in a 3-D ovarian tissue culture system.
Collapse
|
9
|
Liu Y, Mahara A, Kambe Y, Hsu YI, Yamaoka T. Endothelial cell adhesion and blood response to hemocompatible peptide 1 (HCP-1), REDV, and RGD peptide sequences with free N-terminal amino groups immobilized on a biomedical expanded polytetrafluorethylene surface. Biomater Sci 2021; 9:1034-1043. [PMID: 33336665 DOI: 10.1039/d0bm01396j] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Blood compatibility generally requires two contradictory characteristics: reduced protein/platelet adhesion and excellent endothelium-related cell affinity. To understand the effect of cell adhesion peptides on blood compatibility, the peptides REDV, RGD, and hemocompatible peptide-1 (HCP-1) were immobilized on an expanded polytetrafluorethylene (ePTFE) surface and evaluated in vitro, in situ, and in vivo. Since the terminal amino groups of functional peptides often have an important effect, a cysteine residue was added to the C terminal and used for immobilization to keep the terminal amino groups free. Maleimide groups were added to carboxylic groups of highly hydrophilic and biologically inert (bioinert) polymer chains grafted onto ePTFE and coupled with cysteine residues. In vitro tests revealed that free N-terminal HCP-1 and RGD-immobilized surfaces improved the adhesion and spread of human umbilical vein endothelial cells (HUVECs), while, unexpectedly, a free N-terminal adjacent to REDV suppressed cell affinity. In situ evaluation with a porcine closed-circuit system for 2 h showed that no platelets adhered to the modified ePTFE sutures due to the bioinert graft chain containing phosphorylcholine groups. Simultaneously, leukocyte-related and endothelium-related cells were observed on RGD-immobilized ePTFE sutures because RGD was recognized by broad types of cells. These cells were not observed on the HCP-1- and REDV-immobilized ePTFE sutures, which may be due to insufficient exposure time. HCP-1-modified ePTFE graft implantation in a porcine femorofemoral (FF) bypass model for 10 days showed that the thrombus layer was clearly mitigated by HCP-1 immobilization. This study suggests that the HCP-1-immobilized ePTFE surface has potential for long-term application by mitigating thrombus and supporting endothelial cell adhesion.
Collapse
Affiliation(s)
- Yihua Liu
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan.
| | - Atsushi Mahara
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan.
| | - Yusuke Kambe
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan.
| | - Yu-I Hsu
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan.
| | - Tetsuji Yamaoka
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan.
| |
Collapse
|
10
|
Ludwig BS, Kessler H, Kossatz S, Reuning U. RGD-Binding Integrins Revisited: How Recently Discovered Functions and Novel Synthetic Ligands (Re-)Shape an Ever-Evolving Field. Cancers (Basel) 2021; 13:1711. [PMID: 33916607 PMCID: PMC8038522 DOI: 10.3390/cancers13071711] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 12/19/2022] Open
Abstract
Integrins have been extensively investigated as therapeutic targets over the last decades, which has been inspired by their multiple functions in cancer progression, metastasis, and angiogenesis as well as a continuously expanding number of other diseases, e.g., sepsis, fibrosis, and viral infections, possibly also Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2). Although integrin-targeted (cancer) therapy trials did not meet the high expectations yet, integrins are still valid and promising targets due to their elevated expression and surface accessibility on diseased cells. Thus, for the future successful clinical translation of integrin-targeted compounds, revisited and innovative treatment strategies have to be explored based on accumulated knowledge of integrin biology. For this, refined approaches are demanded aiming at alternative and improved preclinical models, optimized selectivity and pharmacological properties of integrin ligands, as well as more sophisticated treatment protocols considering dose fine-tuning of compounds. Moreover, integrin ligands exert high accuracy in disease monitoring as diagnostic molecular imaging tools, enabling patient selection for individualized integrin-targeted therapy. The present review comprehensively analyzes the state-of-the-art knowledge on the roles of RGD-binding integrin subtypes in cancer and non-cancerous diseases and outlines the latest achievements in the design and development of synthetic ligands and their application in biomedical, translational, and molecular imaging approaches. Indeed, substantial progress has already been made, including advanced ligand designs, numerous elaborated pre-clinical and first-in-human studies, while the discovery of novel applications for integrin ligands remains to be explored.
Collapse
Affiliation(s)
- Beatrice S. Ludwig
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
| | - Horst Kessler
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Susanne Kossatz
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Ute Reuning
- Clinical Research Unit, Department of Obstetrics and Gynecology, University Hospital Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany
| |
Collapse
|
11
|
Ayo A, Laakkonen P. Peptide-Based Strategies for Targeted Tumor Treatment and Imaging. Pharmaceutics 2021; 13:pharmaceutics13040481. [PMID: 33918106 PMCID: PMC8065807 DOI: 10.3390/pharmaceutics13040481] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 02/03/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide. The development of cancer-specific diagnostic agents and anticancer toxins would improve patient survival. The current and standard types of medical care for cancer patients, including surgery, radiotherapy, and chemotherapy, are not able to treat all cancers. A new treatment strategy utilizing tumor targeting peptides to selectively deliver drugs or applicable active agents to solid tumors is becoming a promising approach. In this review, we discuss the different tumor-homing peptides discovered through combinatorial library screening, as well as native active peptides. The different structure–function relationship data that have been used to improve the peptide’s activity and conjugation strategies are highlighted.
Collapse
Affiliation(s)
- Abiodun Ayo
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
| | - Pirjo Laakkonen
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
- Laboratory Animal Center, HiLIFE—Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
- Correspondence: ; Tel.: +358-50-4489100
| |
Collapse
|
12
|
Mezu-Ndubuisi OJ, Maheshwari A. The role of integrins in inflammation and angiogenesis. Pediatr Res 2021; 89:1619-1626. [PMID: 33027803 PMCID: PMC8249239 DOI: 10.1038/s41390-020-01177-9] [Citation(s) in RCA: 218] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/18/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023]
Abstract
Integrins are heterodimeric transmembrane cell adhesion molecules made up of alpha (α) and beta (β) subunits arranged in numerous dimeric pairings. These complexes have varying affinities to extracellular ligands. Integrins regulate cellular growth, proliferation, migration, signaling, and cytokine activation and release and thereby play important roles in cell proliferation and migration, apoptosis, tissue repair, as well as in all processes critical to inflammation, infection, and angiogenesis. This review presents current evidence from human and animal studies on integrin structure and molecular signaling, with particular emphasis on signal transduction in infants. We have included evidence from our own laboratory studies and from an extensive literature search in databases PubMed, EMBASE, Scopus, and the electronic archives of abstracts presented at the annual meetings of the Pediatric Academic Societies. To avoid bias in identification of existing studies, key words were short-listed prior to the actual search both from anecdotal experience and from PubMed's Medical Subject Heading (MeSH) thesaurus. IMPACT: Integrins are a family of ubiquitous αβ heterodimeric receptors that interact with numerous ligands in physiology and disease. Integrins play a key role in cell proliferation, tissue repair, inflammation, infection, and angiogenesis. This review summarizes current evidence from human and animal studies on integrin structure and molecular signaling and promising role in diseases of inflammation, infection, and angiogenesis in infants. This review shows that integrin receptors and ligands are novel therapeutic targets of clinical interest and hold promise as novel therapeutic targets in the management of several neonatal diseases.
Collapse
Affiliation(s)
- Olachi J. Mezu-Ndubuisi
- grid.14003.360000 0001 2167 3675Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
| | - Akhil Maheshwari
- grid.21107.350000 0001 2171 9311Department of Pediatrics, Johns Hopkins University, Baltimore, MD USA
| |
Collapse
|
13
|
Quantitative proteomic analysis of human plasma using tandem mass tags to identify novel biomarkers for herpes zoster. J Proteomics 2020; 225:103879. [PMID: 32585426 DOI: 10.1016/j.jprot.2020.103879] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 11/21/2022]
Abstract
Herpes zoster (HZ), commonly called shingles, it is a distinctive syndrome caused by reactivation of varicella zoster virus (VZV). A better understanding of the biological characteristics of HZ patients can help develop new targeted therapies to improve the prognosis. High-throughput proteomics technology can deeply study the molecular changes in the development and progression of HZ disease and integrate different levels of information, this is important to help make clinical decisions. Circulating blood contains a lot of biological information, we conducted a proteomics study of patient plasma, hoping to identify key proteins that could indicate the development of HZ. Compared to healthy human plasma, we found 44 differentially expressed proteins in the plasma of HZ patients, the main pathways involved in these molecules are MAPK signaling pathway, Neuroactive ligand-receptor interaction, Acute myeloid leukemia, Transcriptional misregulation in cancer. We found that 27 proteins have direct protein-protein interactions. Based on the comprehensive score, we identified six key molecules as candidate molecules for further study, and then validated another 80 plasma samples (40 HZ patient plasma and 40 healthy human plasma) using enzyme-linked immunosorbent assay (ELISA), immunoblot assay and receiver operating characteristic (ROC) curve analysis. Finally, we found that the expression levels of these three proteins (PLG, F2, VTN) were significantly lower than those of healthy controls (P < .05). To the best of our knowledge, we first used tandem mass tag (TMT) combined with liquid chromatography-mass spectrometry (LC-MS/MS) to screen for differentially expressed proteins in plasma between HZ patients and healthy individuals. It is preliminarily proved that the plasma protein expression profile of HZ patients is different from that of uninfected patients, it has also been found that these three altered key proteins may be used as biomarkers to test early HZ infection. This study reveals new insights into HZ that help to more accurately identify early HZ patients and to find new therapeutic targets. SIGNIFICANCE: Varicella-zoster virus (VZV; termed human alphaherpesvirus 3 by the International Committee on Taxonomy of Viruses) is a herpesvirus that is ubiquitous in humans and can cause chickenpox and herpes zoster (HZ). After the initial infection of varicella, the VZV goes into a dormant state in the sensory ganglia and cranial nerves. As age or immunosuppression increases, the cellular immunity to VZV decreases, and the virus reactivates and spreads along the sensory nerves to the skin, causing a unique prodromal pain followed by a rash. About one in five people around the world may be infected with VZV at some point in their lives. According to statistics, about one-third of infected people will develop HZ in their lifetime, and an estimated 1 million cases of herpes zoster occur in the United States each year. Herpes zoster can occur at any age and is usually less severe in children and young adults, but the greatest morbidity and mortality are observed in elderly and immunocompromised patients. 20% of patients with HZ have complications including vasculitis, increased risk of myocardial infarction, or postherpetic neuralgia, the overall mortality rate of patients with HZ in the United States is close to 5%. Considering the wide clinical severity and complications of this disease, there is a great need for biomarkers that contribute to early diagnosis, classification of risks, and prediction of outcomes, which will help elucidate the mechanisms underlying their clinical development. As a useful tool in biology, quantitative proteomics can repeatedly identify and accurately quantify proteins in a variety of biological samples. Proteomic analysis focuses on translational proteins, which play a direct role in most biological processes. Although a small number of proteins can be studied simultaneously with traditional methods, such as ELISA and Western blotting, typical proteomics studies can simultaneously analyze thousands of proteins for a more comprehensive identification. Proteomics has been successfully applied to human-based disease research, Analysis of exposed and unexposed subjects based on mass spectrometry (MS) has been found to reveal altered expression of proteins that can be identified as intermediate biomarkers of early disease effects. Tandem mass tags (TMTs) are chemical labels used for MS-based identification and quantification of biological molecules. TMTs play an important role in proteomic analysis in a variety of samples such as cells, tissues, and body fluids. The body fluids that are often detected clinically are blood, which are easy to obtain and contain abundant biological information related to physiological and pathological processes, we hope to develop protein biomarkers from these blood. Therefore, in order to better characterize the pathological process of HZ patients, we performed proteomic analysis of HZ patients and healthy human plasma using the TMT method. This comparison aims to identify specific processes in the development of HZ disease through protein profiling, which may help to improve our biological understanding of HZ.
Collapse
|
14
|
Wu PH, Opadele AE, Onodera Y, Nam JM. Targeting Integrins in Cancer Nanomedicine: Applications in Cancer Diagnosis and Therapy. Cancers (Basel) 2019; 11:E1783. [PMID: 31766201 PMCID: PMC6895796 DOI: 10.3390/cancers11111783] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 02/08/2023] Open
Abstract
Due to advancements in nanotechnology, the application of nanosized materials (nanomaterials) in cancer diagnostics and therapeutics has become a leading area in cancer research. The decoration of nanomaterial surfaces with biological ligands is a major strategy for directing the actions of nanomaterials specifically to cancer cells. These ligands can bind to specific receptors on the cell surface and enable nanomaterials to actively target cancer cells. Integrins are one of the cell surface receptors that regulate the communication between cells and their microenvironment. Several integrins are overexpressed in many types of cancer cells and the tumor microvasculature and function in the mediation of various cellular events. Therefore, the surface modification of nanomaterials with integrin-specific ligands not only increases their binding affinity to cancer cells but also enhances the cellular uptake of nanomaterials through the intracellular trafficking of integrins. Moreover, the integrin-specific ligands themselves interfere with cancer migration and invasion by interacting with integrins, and this finding provides a novel direction for new treatment approaches in cancer nanomedicine. This article reviews the integrin-specific ligands that have been used in cancer nanomedicine and provides an overview of the recent progress in cancer diagnostics and therapeutic strategies involving the use of integrin-targeted nanomaterials.
Collapse
Affiliation(s)
- Ping-Hsiu Wu
- Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 060-8638, Hokkaido, Japan
| | - Abayomi Emmanuel Opadele
- Molecular and Cellular Dynamics Research, Graduate School of Biomedical Science and Engineering, Hokkaido University, Sapporo 060-8638, Hokkaido, Japan;
| | - Yasuhito Onodera
- Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 060-8638, Hokkaido, Japan
- Department of Molecular Biology, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Hokkaido, Japan
| | - Jin-Min Nam
- Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 060-8638, Hokkaido, Japan
| |
Collapse
|
15
|
Shin K, Lechtenberg BC, Fujimoto LM, Yao Y, Bartra SS, Plano GV, Marassi FM. Structure of human Vitronectin C-terminal domain and interaction with Yersinia pestis outer membrane protein Ail. SCIENCE ADVANCES 2019; 5:eaax5068. [PMID: 31535027 PMCID: PMC6739113 DOI: 10.1126/sciadv.aax5068] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 08/08/2019] [Indexed: 05/27/2023]
Abstract
Vitronectin (Vn) is a major component of blood that controls many processes central to human biology. It is a drug target and a key factor in cell and tissue engineering applications, but despite long-standing efforts, little is known about the molecular basis for its functions. Here, we define the domain organization of Vn, report the crystal structure of its carboxyl-terminal domain, and show that it harbors the binding site for the Yersinia pestis outer membrane protein Ail, which recruits Vn to the bacterial cell surface to evade human host defenses. Vn forms a single four-bladed β/α-propeller that serves as a hub for multiple functions. The structure explains key features of native Vn and provides a blueprint for understanding and targeting this essential human protein.
Collapse
Affiliation(s)
- Kyungsoo Shin
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Bernhard C. Lechtenberg
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Lynn M. Fujimoto
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Yong Yao
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Sara Schesser Bartra
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | - Gregory V. Plano
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | - Francesca M. Marassi
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
16
|
Mahzoon S, Detamore MS. Chondroinductive Peptides: Drawing Inspirations from Cell–Matrix Interactions. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:249-257. [DOI: 10.1089/ten.teb.2018.0003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Salma Mahzoon
- School of Aerospace and Mechanical Engineering, University of Oklahoma, Norman, Oklahoma
| | - Michael S. Detamore
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma
| |
Collapse
|
17
|
Tam RY, Yockell-Lelièvre J, Smith LJ, Julian LM, Baker AEG, Choey C, Hasim MS, Dimitroulakos J, Stanford WL, Shoichet MS. Rationally Designed 3D Hydrogels Model Invasive Lung Diseases Enabling High-Content Drug Screening. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1806214. [PMID: 30589121 DOI: 10.1002/adma.201806214] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/09/2018] [Indexed: 06/09/2023]
Abstract
Cell behavior is highly dependent upon microenvironment. Thus, to identify drugs targeting metastatic cancer, screens need to be performed in tissue mimetic substrates that allow cell invasion and matrix remodeling. A novel biomimetic 3D hydrogel platform that enables quantitative analysis of cell invasion and viability at the individual cell level is developed using automated data acquisition methods with an invasive lung disease (lymphangioleiomyomatosis, LAM) characterized by hyperactive mammalian target of rapamycin complex 1 (mTORC1) signaling as a model. To test the lung-mimetic hydrogel platform, a kinase inhibitor screen is performed using tuberous sclerosis complex 2 (TSC2) hypomorphic cells, identifying Cdk2 inhibition as a putative LAM therapeutic. The 3D hydrogels mimic the native niche, enable multiple modes of invasion, and delineate phenotypic differences between healthy and diseased cells, all of which are critical to effective drug screens of highly invasive diseases including lung cancer.
Collapse
Affiliation(s)
- Roger Y Tam
- Department of Chemical Engineering and Applied Chemistry, Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, K1H 8L6, Canada
| | - Julien Yockell-Lelièvre
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, K1H 8L6, Canada
| | - Laura J Smith
- Department of Chemical Engineering and Applied Chemistry, Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Lisa M Julian
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, K1H 8L6, Canada
| | - Alexander E G Baker
- Department of Chemical Engineering and Applied Chemistry, Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Chandarong Choey
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, K1H 8L6, Canada
| | - Mohamed S Hasim
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, K1H 8L6, Canada
| | - Jim Dimitroulakos
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, K1H 8L6, Canada
| | - William L Stanford
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada
| | - Molly S Shoichet
- Department of Chemical Engineering and Applied Chemistry, Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario, M5S 3H6, Canada
| |
Collapse
|
18
|
Cao Y, Li X, Lu C, Zhan X. The relationship between vitronectin and hepatic insulin resistance in type 2 diabetes mellitus. Endocr J 2018; 65:747-753. [PMID: 29780059 DOI: 10.1507/endocrj.ej17-0504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The World Health Organization (WHO) estimates that approximately 300 million people will suffer from diabetes mellitus by 2025. Type 2 diabetes mellitus (T2DM) is much more prevalent. T2DM comprises approximately 90% of diabetes mellitus cases, and it is caused by a combination of insulin resistance and inadequate compensatory insulin secretory response. In this study, we aimed to compare the plasma vitronectin (VN) levels between patients with T2DM and insulin resistance (IR) and healthy controls. Seventy patients with IR and 70 age- and body mass index (BMI)-matched healthy controls were included in the study. The insulin, Waist-to-Hip Ratio (WHR), C-peptide (CP) and VN levels of all participants were examined. The homeostasis model of assessment for insulin resistence index (HOMA-IR (CP)) formula was used to calculate insulin resistance. The levels of BMI, fasting plasma gluose (FPG), 2-hour postprandial glucose (2hPG), glycated hemoglobins (HbA1c), and HOMA-IR (CP) were significantly elevated in case group compared with controls. VN was found to be significantly decreased in case group. (VN Mean (Std): 8.55 (2.92) versus 12.88 (1.26) ng/mL p < 0.001). Multiple linear regression analysis was performed. This model explained 43.42% of the total variability of VN. Multiple linear regression analysis showed that HOMA-IR (CP) and age independently predicted VN levels. The VN may be a candidate target for the appraisal of hepatic insulin resistance in patients with T2DM.
Collapse
Affiliation(s)
- Yan Cao
- Department of Endocrinology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150007, China
| | - Xinyu Li
- Department of Endocrinology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150007, China
- Department of Pharmacology, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Harbin Medical University, Harbin, Hei Long Jiang Province 150081, PR China
| | - Chong Lu
- Department of Neurology, Heilongjiang Provincial Hospital, Harbin, Hei Long Jiang Province 150036, PR China
| | - Xiaorong Zhan
- Department of Endocrinology, the First Affiliated Hospital of Harbin Medical University, Harbin, 150007, China
| |
Collapse
|
19
|
Keasey MP, Jia C, Pimentel LF, Sante RR, Lovins C, Hagg T. Blood vitronectin is a major activator of LIF and IL-6 in the brain through integrin-FAK and uPAR signaling. J Cell Sci 2018; 131:jcs.202580. [PMID: 29222114 DOI: 10.1242/jcs.202580] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 12/06/2017] [Indexed: 12/18/2022] Open
Abstract
We defined how blood-derived vitronectin (VTN) rapidly and potently activates leukemia inhibitory factor (LIF) and pro-inflammatory interleukin 6 (IL-6) in vitro and after vascular injury in the brain. Treatment with VTN (but not fibrinogen, fibronectin, laminin-111 or collagen-I) substantially increased LIF and IL-6 within 4 h in C6-astroglioma cells, while VTN-/- mouse plasma was less effective than that from wild-type mice. LIF and IL-6 were induced by intracerebral injection of recombinant human (rh)VTN in mice, but induction seen upon intracerebral hemorrhage was less in VTN-/- mice than in wild-type littermates. In vitro, VTN effects were inhibited by RGD, αvβ3 and αvβ5 integrin-blocking peptides and antibodies. VTN activated focal adhesion kinase (FAK; also known as PTK2), whereas pharmacological- or siRNA-mediated inhibition of FAK, but not PYK2, reduced the expression of LIF and IL-6 in C6 and endothelial cells and after traumatic cell injury. Dominant-negative FAK (Y397F) reduced the amount of injury-induced LIF and IL-6. Pharmacological inhibition or knockdown of uPAR (also known as PLAUR), which binds VTN, also reduced cytokine expression, possibly through a common target of uPAR and integrins. We propose that VTN leakage into tissues promotes inflammation. Integrin-FAK signaling is therefore a novel IL-6 and LIF regulation mechanism relevant to the inflammation and stem cell fields.
Collapse
Affiliation(s)
- Matthew P Keasey
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Cuihong Jia
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Lylyan F Pimentel
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA.,Keizo Asami Laboratory (LIKA), Universidade Federal de Pernambuco, Recife, PE, Brasil
| | - Richard R Sante
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Chiharu Lovins
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Theo Hagg
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| |
Collapse
|
20
|
Long-Term Maintenance of Human Pluripotent Stem Cells on cRGDfK-Presenting Synthetic Surfaces. Sci Rep 2018; 8:701. [PMID: 29335618 PMCID: PMC5768753 DOI: 10.1038/s41598-018-19209-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 12/27/2017] [Indexed: 12/12/2022] Open
Abstract
Synthetic human pluripotent stem cell (hPSC) culture surfaces with defined physical and chemical properties will facilitate improved research and therapeutic applications of hPSCs. In this study, synthetic surfaces for hPSC culture in E8 medium were produced for screening by modifying two polymer brush coatings [poly(acrylamide-co-acrylic acid) (PAAA) and poly(acrylamide-co-propargyl acrylamide) (PAPA)] to present single peptides. Adhesion of hPSC colonies was more consistently observed on surfaces modified with cRGDfK compared to surfaces modified with other peptide sequences tested. PAPA-coated polystyrene flasks with coupled cRGDfK (cRGDfK-PAPA) were then used for long-term studies of three hPSC lines (H9, hiPS-NHF1.3, Genea-02). Cell lines maintained for ten passages on cRGDfK-PAPA were assessed for colony morphology, proliferation rate, maintenance of OCT4 expression, cell viability at harvest, teratoma formation potential, and global gene expression as assessed by the PluriTest™ assay. cRGDfK-PAPA and control cultures maintained on Geltrex™ produced comparable results in most assays. No karyotypic abnormalities were detected in cultures maintained on cRGDfK-PAPA, while abnormalities were detected in cultures maintained on Geltrex™, StemAdhere™ or Synthemax™. This is the first report of long term maintenance of hPSC cultures on the scalable, stable, and cost-effective cRGDfK-PAPA coating.
Collapse
|
21
|
Nickel J, Ten Dijke P, Mueller TD. TGF-β family co-receptor function and signaling. Acta Biochim Biophys Sin (Shanghai) 2018; 50:12-36. [PMID: 29293886 DOI: 10.1093/abbs/gmx126] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 11/08/2017] [Indexed: 01/04/2023] Open
Abstract
Transforming growth factor-β (TGF-β) family members, which include TGF-βs, activins and bone morphogenetic proteins, are pleiotropic cytokines that elicit cell type-specific effects in a highly context-dependent manner in many different tissues. These secreted protein ligands signal via single-transmembrane Type I and Type II serine/threonine kinase receptors and intracellular SMAD transcription factors. Deregulation in signaling has been implicated in a broad array of diseases, and implicate the need for intricate fine tuning in cellular signaling responses. One important emerging mechanism by which TGF-β family receptor signaling intensity, duration, specificity and diversity are regulated and/or mediated is through cell surface co-receptors. Here, we provide an overview of the co-receptors that have been identified for TGF-β family members. While some appear to be specific to TGF-β family members, others are shared with other pathways and provide possible ways for signal integration. This review focuses on novel functions of TGF-β family co-receptors, which continue to be discovered.
Collapse
Affiliation(s)
- Joachim Nickel
- Universitätsklinikum Würzburg, Lehrstuhl für Tissue Engineering und Regenerative Medizin und Fraunhofer Institut für Silicatforschung (ISC), Translationszentrum "Regenerative Therapien", Röntgenring 11, D-97070 Würzburg, Germany
| | - Peter Ten Dijke
- Department of Molecular and Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, The Netherlands
| | - Thomas D Mueller
- Lehrstuhl für molekulare Pflanzenphysiologie und Biophysik, Julius-von-Sachs Institut für Biowissenschaften, Universität Würzburg, Julius-von-Sachs-Platz 2, D-97082 Würzburg, Germany
| |
Collapse
|
22
|
αvβ3 and α5β1 integrin-specific ligands: From tumor angiogenesis inhibitors to vascularization promoters in regenerative medicine? Biotechnol Adv 2017; 36:208-227. [PMID: 29155160 DOI: 10.1016/j.biotechadv.2017.11.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/07/2017] [Accepted: 11/13/2017] [Indexed: 12/30/2022]
Abstract
Integrins are cell adhesion receptors predominantly important during normal and tumor angiogenesis. A sequence present on several extracellular matrix proteins composed of Arg-Gly-Asp (RGD) has attracted attention due to its role in cell adhesion mediated by integrins. The development of ligands that can bind to integrins involved in tumor angiogenesis and brake disease progression has resulted in new investigational drug entities reaching the clinical trial phase in humans. The use of integrin-specific ligands can be useful for the vascularization of regenerative medicine constructs, which remains a major limitation for translation into clinical practice. In order to enhance vascularization, immobilization of integrin-specific RGD peptidomimetics within constructs is a recommended approach, due to their high specificity and selectivity towards certain desired integrins. This review endeavours to address the potential of peptidomimetic-coated biomaterials as vascular network promoters for regenerative medicine purposes. Clinical studies involving molecules tracking active integrins in cancer angiogenesis and reasons for their failure are also addressed.
Collapse
|
23
|
Exploring the Role of RGD-Recognizing Integrins in Cancer. Cancers (Basel) 2017; 9:cancers9090116. [PMID: 28869579 PMCID: PMC5615331 DOI: 10.3390/cancers9090116] [Citation(s) in RCA: 313] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/28/2017] [Accepted: 08/31/2017] [Indexed: 12/18/2022] Open
Abstract
Integrins are key regulators of communication between cells and with their microenvironment. Eight members of the integrin superfamily recognize the tripeptide motif Arg-Gly-Asp (RGD) within extracelluar matrix (ECM) proteins. These integrins constitute an important subfamily and play a major role in cancer progression and metastasis via their tumor biological functions. Such transmembrane adhesion and signaling receptors are thus recognized as promising and well accessible targets for novel diagnostic and therapeutic applications for directly attacking cancer cells and their fatal microenvironment. Recently, specific small peptidic and peptidomimetic ligands as well as antibodies binding to distinct integrin subtypes have been developed and synthesized as new drug candidates for cancer treatment. Understanding the distinct functions and interplay of integrin subtypes is a prerequisite for selective intervention in integrin-mediated diseases. Integrin subtype-specific ligands labelled with radioisotopes or fluorescent molecules allows the characterization of the integrin patterns in vivo and later the medical intervention via subtype specific drugs. The coating of nanoparticles, larger proteins, or encapsulating agents by integrin ligands are being explored to guide cytotoxic reagents directly to the cancer cell surface. These ligands are currently under investigation in clinical studies for their efficacy in interference with tumor cell adhesion, migration/invasion, proliferation, signaling, and survival, opening new treatment approaches in personalized medicine.
Collapse
|
24
|
Varun D, Srinivasan GR, Tsai YH, Kim HJ, Cutts J, Petty F, Merkley R, Stephanopoulos N, Dolezalova D, Marsala M, Brafman DA. A robust vitronectin-derived peptide for the scalable long-term expansion and neuronal differentiation of human pluripotent stem cell (hPSC)-derived neural progenitor cells (hNPCs). Acta Biomater 2017; 48:120-130. [PMID: 27989923 DOI: 10.1016/j.actbio.2016.10.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 10/03/2016] [Accepted: 10/26/2016] [Indexed: 12/22/2022]
Abstract
Despite therapeutic advances, neurodegenerative diseases and disorders remain some of the leading causes of mortality and morbidity in the United States. Therefore, cell-based therapies to replace lost or damaged neurons and supporting cells of the central nervous system (CNS) are of great therapeutic interest. To that end, human pluripotent stem cell (hPSC) derived neural progenitor cells (hNPCs) and their neuronal derivatives could provide the cellular 'raw material' needed for regenerative medicine therapies for a variety of CNS disorders. In addition, hNPCs derived from patient-specific hPSCs could be used to elucidate the underlying mechanisms of neurodegenerative diseases and identify potential drug candidates. However, the scientific and clinical application of hNPCs requires the development of robust, defined, and scalable substrates for their long-term expansion and neuronal differentiation. In this study, we rationally designed a vitronectin-derived peptide (VDP) that served as an adhesive growth substrate for the long-term expansion of several hNPC lines. Moreover, VDP-coated surfaces allowed for the directed neuronal differentiation of hNPC at levels similar to cells differentiated on traditional extracellular matrix protein-based substrates. Overall, the ability of VDP to support the long-term expansion and directed neuronal differentiation of hNPCs will significantly advance the future translational application of these cells in treating injuries, disorders, and diseases of the CNS.
Collapse
|
25
|
Komazawa H, Saiki I, Igarashi YU, Azuma I, Kojima M, Orikasa A, Ono M, Itoh I. Inhibition of Tumor Metastasis by a Synthetic Polymer Containing a Cell-Adhesive RGDS Peptide. J BIOACT COMPAT POL 2016. [DOI: 10.1177/088391159300800305] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A water soluble polymer containing cell adhesive Arg-Gly-Asp- Ser (RGDS) sequence, i.e., poly(carboxyethylmethacrylamide-RGDS) conjugate [poly(CEMA-RGDS)], was synthesized. Poly(CEMA-RGDS) inhibited lung metastasis of B16-BL6 melanoma cells in a dose-dependent manner (20-1000 μg) and liver metastasis of L5178YML25 lymphoma cells. A mixture of poly- (CEMA) and RGDS peptide or poly(CEMA) alone did not show any inhibitory effect on lung metastasis. The Gly-Arg-Gly-Asp-Ser (GRGDS) peptide required high doses (3000 μ g) to obtain a sufficient antimetastatic effect. An in vitro study showed that poly(CEMA-RGDS) as well as RGDS + poly(CEMA) gave similar inhibition of B16-BL6 cell invasion into reconstituted basement mem brane Matrigel. Intermittent i.v. administration of poly(CEMA-RGDS) after in oculation with B16-BL6 cells caused significant inhibition of spontaneous lung metastasis as compared with the multiple administration of RGDS, poly- (CEMA) or untreated control. These results demonstrate that the conjugation
Collapse
Affiliation(s)
- Hiroyuki Komazawa
- Institute of Immunological Science Hokkaido University Kita-15, Nishi-7 Kita-ku, Sapporo 060, Japan
| | - Ikuo Saiki
- Institute of Immunological Science Hokkaido University Kita-15, Nishi-7 Kita-ku, Sapporo 060, Japan
| | - YU Igarashi
- Institute of Immunological Science Hokkaido University Kita-15, Nishi-7 Kita-ku, Sapporo 060, Japan
| | - Ichiro Azuma
- Institute of Immunological Science Hokkaido University Kita-15, Nishi-7 Kita-ku, Sapporo 060, Japan
| | - Masayoshi Kojima
- Fuji Photo Film Co., Ltd. Ashigara Research Laboratories 210 Nakanuma, Minamiashigara, 250-01, Japan
| | - Atsushi Orikasa
- Fuji Photo Film Co., Ltd. Ashigara Research Laboratories 210 Nakanuma, Minamiashigara, 250-01, Japan
| | - Mitsunori Ono
- Fuji Photo Film Co., Ltd. Ashigara Research Laboratories 210 Nakanuma, Minamiashigara, 250-01, Japan
| | - Isamu Itoh
- Fuji Photo Film Co., Ltd. Ashigara Research Laboratories 210 Nakanuma, Minamiashigara, 250-01, Japan
| |
Collapse
|
26
|
Mas-Moruno C, Fraioli R, Rechenmacher F, Neubauer S, Kapp TG, Kessler H. αvβ3- or α5β1-Integrin-Selective Peptidomimetics for Surface Coating. Angew Chem Int Ed Engl 2016; 55:7048-67. [PMID: 27258759 DOI: 10.1002/anie.201509782] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Indexed: 12/21/2022]
Abstract
Engineering biomaterials with integrin-binding activity is a very powerful approach to promote cell adhesion, modulate cell behavior, and induce specific biological responses at the surface level. The aim of this Review is to illustrate the evolution of surface-coating molecules in this field: from peptides and proteins with relatively low integrin-binding activity and receptor selectivity to highly active and selective peptidomimetic ligands. In particular, we will bring into focus the difficult challenge of achieving selectivity between the two closely related integrin subtypes αvβ3 and α5β1. The functionalization of surfaces with such peptidomimetics opens the way for a new generation of highly specific cell-instructive surfaces to dissect the biological role of integrin subtypes and for application in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Carlos Mas-Moruno
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering and Centre for Research in NanoEngineering, Universitat Politècnica de Catalunya (UPC), Diagonal 647, 08028, Barcelona, Spain.
| | - Roberta Fraioli
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering and Centre for Research in NanoEngineering, Universitat Politècnica de Catalunya (UPC), Diagonal 647, 08028, Barcelona, Spain
| | - Florian Rechenmacher
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85748, Garching, Germany
| | - Stefanie Neubauer
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85748, Garching, Germany
| | - Tobias G Kapp
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85748, Garching, Germany
| | - Horst Kessler
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85748, Garching, Germany.
| |
Collapse
|
27
|
Mas-Moruno C, Fraioli R, Rechenmacher F, Neubauer S, Kapp TG, Kessler H. αvβ3- oder α5β1-Integrin-selektive Peptidmimetika für die Oberflächenbeschichtung. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201509782] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Carlos Mas-Moruno
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering and Centre for Research in NanoEngineering; Universitat Politècnica de Catalunya (UPC); Diagonal 647 08028 Barcelona Spanien
| | - Roberta Fraioli
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering and Centre for Research in NanoEngineering; Universitat Politècnica de Catalunya (UPC); Diagonal 647 08028 Barcelona Spanien
| | - Florian Rechenmacher
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM); Technische Universität München; Lichtenbergstraße 4 85748 Garching Deutschland
| | - Stefanie Neubauer
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM); Technische Universität München; Lichtenbergstraße 4 85748 Garching Deutschland
| | - Tobias G. Kapp
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM); Technische Universität München; Lichtenbergstraße 4 85748 Garching Deutschland
| | - Horst Kessler
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM); Technische Universität München; Lichtenbergstraße 4 85748 Garching Deutschland
| |
Collapse
|
28
|
Gilad Y, Firer M, Gellerman G. Recent Innovations in Peptide Based Targeted Drug Delivery to Cancer Cells. Biomedicines 2016; 4:E11. [PMID: 28536378 PMCID: PMC5344250 DOI: 10.3390/biomedicines4020011] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 05/16/2016] [Accepted: 05/23/2016] [Indexed: 12/21/2022] Open
Abstract
Targeted delivery of chemotherapeutics and diagnostic agents conjugated to carrier ligands has made significant progress in recent years, both in regards to the structural design of the conjugates and their biological effectiveness. The goal of targeting specific cell surface receptors through structural compatibility has encouraged the use of peptides as highly specific carriers as short peptides are usually non-antigenic, are structurally simple and synthetically diverse. Recent years have seen many developments in the field of peptide based drug conjugates (PDCs), particularly for cancer therapy, as their use aims to bypass off-target side-effects, reducing the morbidity common to conventional chemotherapy. However, no PDCs have as yet obtained regulatory approval. In this review, we describe the evolution of the peptide-based strategy for targeted delivery of chemotherapeutics and discuss recent innovations in the arena that should lead in the near future to their clinical application.
Collapse
Affiliation(s)
- Yosi Gilad
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel.
- Department of Chemical Engineering and Biotechnology, Ariel University, Ariel 40700, Israel.
| | - Michael Firer
- Department of Chemical Engineering and Biotechnology, Ariel University, Ariel 40700, Israel.
| | - Gary Gellerman
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel.
| |
Collapse
|
29
|
Clevenger TN, Hinman CR, Ashley Rubin RK, Smither K, Burke DJ, Hawker CJ, Messina D, Van Epps D, Clegg DO. Vitronectin-Based, Biomimetic Encapsulating Hydrogel Scaffolds Support Adipogenesis of Adipose Stem Cells. Tissue Eng Part A 2016; 22:597-609. [PMID: 26956095 DOI: 10.1089/ten.tea.2015.0550] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Soft tissue defects are relatively common, yet currently used reconstructive treatments have varying success rates, and serious potential complications such as unpredictable volume loss and reabsorption. Human adipose-derived stem cells (ASCs), isolated from liposuction aspirate have great potential for use in soft tissue regeneration, especially when combined with a supportive scaffold. To design scaffolds that promote differentiation of these cells down an adipogenic lineage, we characterized changes in the surrounding extracellular environment during adipogenic differentiation. We found expression changes in both extracellular matrix proteins, including increases in expression of collagen-IV and vitronectin, as well as changes in the integrin expression profile, with an increase in expression of integrins such as αVβ5 and α1β1. These integrins are known to specifically interact with vitronectin and collagen-IV, respectively, through binding to an Arg-Gly-Asp (RGD) sequence. When three different short RGD-containing peptides were incorporated into three-dimensional (3D) hydrogel cultures, it was found that an RGD-containing peptide derived from vitronectin provided strong initial attachment, maintained the desired morphology, and created optimal conditions for in vitro 3D adipogenic differentiation of ASCs. These results describe a simple, nontoxic encapsulating scaffold, capable of supporting the survival and desired differentiation of ASCs for the treatment of soft tissue defects.
Collapse
Affiliation(s)
- Tracy N Clevenger
- 1 Center for Stem Cell Biology and Engineering, University of California , Santa Barbara, Santa Barbara, California.,2 Department of Molecular, Cellular and Developmental Biology, University of California , Santa Barbara, Santa Barbara, California
| | - Cassidy R Hinman
- 1 Center for Stem Cell Biology and Engineering, University of California , Santa Barbara, Santa Barbara, California
| | - Rebekah K Ashley Rubin
- 1 Center for Stem Cell Biology and Engineering, University of California , Santa Barbara, Santa Barbara, California.,2 Department of Molecular, Cellular and Developmental Biology, University of California , Santa Barbara, Santa Barbara, California
| | | | - Daniel J Burke
- 4 Materials Research Laboratory, University of California , Santa Barbara
| | - Craig J Hawker
- 4 Materials Research Laboratory, University of California , Santa Barbara
| | | | | | - Dennis O Clegg
- 1 Center for Stem Cell Biology and Engineering, University of California , Santa Barbara, Santa Barbara, California.,2 Department of Molecular, Cellular and Developmental Biology, University of California , Santa Barbara, Santa Barbara, California
| |
Collapse
|
30
|
Identification and therapeutic potential of a vitronectin binding region of meningococcal msf. PLoS One 2015; 10:e0124133. [PMID: 25826209 PMCID: PMC4380367 DOI: 10.1371/journal.pone.0124133] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 03/13/2015] [Indexed: 01/06/2023] Open
Abstract
The human pathogen Neisseria meningitides (Nm) attains serum resistance via a number of mechanisms, one of which involves binding to the host complement regulator protein vitronectin. We have shown previously that the Meningococcal surface fibril (Msf), a trimeric autotransporter, binds to the activated form of vitronectin (aVn) to increase Nm survival in human serum. In this study, we aimed to identify the aVn-binding region of Msf to assess its potential as an antigen which can elicit antibodies that block aVn binding and/or possess bactericidal properties. Using several recombinant Msf fragments spanning its surface-exposed region, the smallest aVn-binding recombinants were found to span residues 1-86 and 39-124. The use of further deletion constructs and overlapping recombinant Msf fragments suggested that a region of Msf comprising residues 39-82 may be primarily important for aVn binding and that other regions may also be involved but to a lesser extent. Molecular modelling implicated K66 and K68, conserved in all available Msf sequences, to be involved in the interaction. Recombinant fragments which bound to aVn were able to reduce the survival advantage conveyed by aVn-interaction in serum bactericidal assays. Antibodies raised against one such fragment inhibited aVn binding to Msf. In addition, the antibodies enhanced specific killing of Msf-expressing Nm in a dose-dependent manner. Overall, this study identifies an aVn-binding region of Msf, an adhesin known to impart serum resistance properties to the pathogen; and shows that this region of Msf can elicit antibodies with dual properties which reduce pathogen survival within the host and thus has potential as a vaccine antigen.
Collapse
|
31
|
Salazar-Peláez LM, Abraham T, Herrera AM, Correa MA, Ortega JE, Paré PD, Seow CY. Vitronectin expression in the airways of subjects with asthma and chronic obstructive pulmonary disease. PLoS One 2015; 10:e0119717. [PMID: 25768308 PMCID: PMC4358944 DOI: 10.1371/journal.pone.0119717] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 01/31/2015] [Indexed: 11/30/2022] Open
Abstract
Vitronectin, a multifunctional glycoprotein, is involved in coagulation, inhibition of the formation of the membrane attack complex (MAC), cell adhesion and migration, wound healing, and tissue remodeling. The primary cellular source of vitronectin is hepatocytes; it is not known whether resident cells of airways produce vitronectin, even though the glycoprotein has been found in exhaled breath condensate and bronchoalveolar lavage from healthy subjects and patients with interstitial lung disease. It is also not known whether vitronectin expression is altered in subjects with asthma and COPD. In this study, bronchial tissue from 7 asthmatic, 10 COPD and 14 control subjects was obtained at autopsy and analyzed by immunohistochemistry to determine the percent area of submucosal glands occupied by vitronectin. In a separate set of experiments, quantitative colocalization analysis was performed on tracheobronchial tissue sections obtained from donor lungs (6 asthmatics, 4 COPD and 7 controls). Vitronectin RNA and protein expressions in bronchial surface epithelium were examined in 12 subjects who undertook diagnostic bronchoscopy. Vitronectin was found in the tracheobronchial epithelium from asthmatic, COPD, and control subjects, although its expression was significantly lower in the asthmatic group. Colocalization analysis of 3D confocal images indicates that vitronectin is expressed in the glandular serous epithelial cells and in respiratory surface epithelial cells other than goblet cells. Expression of the 65-kDa vitronectin isoform was lower in bronchial surface epithelium from the diseased subjects. The cause for the decreased vitronectin expression in asthma is not clear, however, the reduced concentration of vitronectin in the epithelial/submucosal layer of airways may be linked to airway remodeling.
Collapse
Affiliation(s)
| | - Thomas Abraham
- Penn State Microscopy Imaging Core, Penn State Hershey College of Medicine, Hershey, Pennsylvania, United States of America
| | | | - Mario A. Correa
- Instituto Nacional de Medicina Legal y Ciencias Forenses, Medellín, Colombia
| | | | - Peter D. Paré
- James Hogg Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Chun Y. Seow
- School of Medicine, Universidad CES, Medellín, Colombia
- James Hogg Research Centre, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
32
|
Lambshead JW, Meagher L, O'Brien C, Laslett AL. Defining synthetic surfaces for human pluripotent stem cell culture. CELL REGENERATION 2013; 2:7. [PMID: 25408879 PMCID: PMC4230363 DOI: 10.1186/2045-9769-2-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 11/19/2013] [Indexed: 12/29/2022]
Abstract
Human pluripotent stem cells (hPSCs) are able to self-renew indefinitely and to differentiate into all adult cell types. hPSCs therefore show potential for application to drug screening, disease modelling and cellular therapies. In order to meet this potential, culture conditions must be developed that are consistent, defined, scalable, free of animal products and that facilitate stable self-renewal of hPSCs. Several culture surfaces have recently been reported to meet many of these criteria although none of them have been widely implemented by the stem cell community due to issues with validation, reliability and expense. Most hPSC culture surfaces have been derived from extracellular matrix proteins (ECMPs) and their cell adhesion molecule (CAM) binding motifs. Elucidating the CAM-mediated cell-surface interactions that are essential for the in vitro maintenance of pluripotency will facilitate the optimisation of hPSC culture surfaces. Reports indicate that hPSC cultures can be supported by cell-surface interactions through certain CAM subtypes but not by others. This review summarises the recent reports of defined surfaces for hPSC culture and focuses on the CAMs and ECMPs involved.
Collapse
Affiliation(s)
- Jack W Lambshead
- CSIRO Materials Science and Engineering, Clayton, Victoria 3168 Australia ; Australian Regenerative Medicine Institute, Monash University, Kragujevac, Victoria 3800 Australia
| | - Laurence Meagher
- CSIRO Materials Science and Engineering, Clayton, Victoria 3168 Australia
| | - Carmel O'Brien
- CSIRO Materials Science and Engineering, Clayton, Victoria 3168 Australia ; Australian Regenerative Medicine Institute, Monash University, Kragujevac, Victoria 3800 Australia
| | - Andrew L Laslett
- CSIRO Materials Science and Engineering, Clayton, Victoria 3168 Australia ; Australian Regenerative Medicine Institute, Monash University, Kragujevac, Victoria 3800 Australia ; Department of Zoology, University of Melbourne, Parkville, Victoria 3101 Australia
| |
Collapse
|
33
|
|
34
|
|
35
|
Higuchi A, Ling QD, Hsu ST, Umezawa A. Biomimetic cell culture proteins as extracellular matrices for stem cell differentiation. Chem Rev 2012; 112:4507-40. [PMID: 22621236 DOI: 10.1021/cr3000169] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, 32001 Taiwan.
| | | | | | | |
Collapse
|
36
|
Titushkin I, Sun S, Cho M. Structure and Biology of the Cellular Environment: The Extracellular Matrix. NANOTECHNOLOGY FOR BIOLOGY AND MEDICINE 2012. [DOI: 10.1007/978-0-387-31296-5_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
37
|
Wirkner M, Weis S, San Miguel V, Álvarez M, Gropeanu RA, Salierno M, Sartoris A, Unger RE, Kirkpatrick CJ, del Campo A. Photoactivatable caged cyclic RGD peptide for triggering integrin binding and cell adhesion to surfaces. Chembiochem 2011; 12:2623-9. [PMID: 22058073 DOI: 10.1002/cbic.201100437] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Indexed: 12/19/2022]
Abstract
We report the synthesis and properties of a photoactivatable caged RGD peptide and its application for phototriggering integrin- and cell-binding to surfaces. We analysed in detail 1) the differences in the integrin-binding affinity of the caged and uncaged forms by quartz crystal microbalance (QCM) studies, 2) the efficiency and yield of the photolytic uncaging reaction, 3) the biocompatibility of the photolysis by-products and irradiation conditions, 4) the possibility of site, temporal and density control of integrin-binding and therefore human cell attachment, and 5) the possibility of in situ generation of cell patterns and cell gradients by controlling the UV exposure. These studies provide a clear picture of the potential and limitations of caged RGD for integrin-mediated cell adhesion and demonstrate the application of this approach to the control and study of cell interactions and responses.
Collapse
Affiliation(s)
- Melanie Wirkner
- Max-Planck-Institut für Polymerforschung, Ackermannweg 10, 55128 Mainz, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
NAKAZAWA CT. Structural Analyses of Silk Fibroin from Wild Silkworms and the Application for Cell Adhesive Materials. KOBUNSHI RONBUNSHU 2011. [DOI: 10.1295/koron.68.540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
39
|
Zustiak SP, Durbal R, Leach JB. Influence of cell-adhesive peptide ligands on poly(ethylene glycol) hydrogel physical, mechanical and transport properties. Acta Biomater 2010; 6:3404-14. [PMID: 20385260 PMCID: PMC2910244 DOI: 10.1016/j.actbio.2010.03.040] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Revised: 03/18/2010] [Accepted: 03/29/2010] [Indexed: 10/19/2022]
Abstract
Synthetic three-dimensional scaffolds for cell and tissue engineering routinely utilize peptide ligands to provide sites for cell adhesion and to promote cellular activity. Given the fact that recent studies have dedicated great attention to the mechanisms by which cell behavior is influenced by various ligands and scaffold material properties, it is surprising that little work to date has been carried out to investigate the influence of covalently bound ligands on hydrogel material properties. Herein we report the influence of three common ligands utilized in tissue engineering, namely RGD, YIGSR and IKVAV, on the mechanical properties of cross-linked poly(ethylene glycol) (PEG) hydrogels. The effect of the ligands on hydrogel storage modulus, swelling ratio, mesh size and also on the diffusivity of bovine serum albumin through the hydrogel were investigated in detail. We identified conditions under which these ligands strikingly influence the properties of the material. The extent of influence and whether the ligand increases or decreases a specific property is linked to ligand type and concentration. Further, we pinpoint mechanisms by which the ligands interact with the PEG network. This work thus provides specific evidence for interactions between peptide ligands and cross-linked PEG hydrogels that have a significant impact on hydrogel material and transport properties. As a result, this work may have important implications for interpreting cell experiments carried out with ligand-modified hydrogels, because the addition of ligand may affect not only the scaffold's biological properties, but also key physical properties of the system.
Collapse
Affiliation(s)
- Silviya P Zustiak
- Department of Chemical and Biochemical Engineering, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | | | | |
Collapse
|
40
|
Beattie J, McIntosh L, van der Walle CF. Cross-talk between the insulin-like growth factor (IGF) axis and membrane integrins to regulate cell physiology. J Cell Physiol 2010; 224:605-11. [PMID: 20432472 DOI: 10.1002/jcp.22183] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The biology of cross-talk between activated growth factor receptors and cell-surface integrins is an area which has attracted much interest in recent years (Schwartz and Ginsberg, 2002). This review discusses the relationship between the insulin-like growth factor (IGF) axis and cell-surface integrin receptors in the regulation of various aspects of cell physiology. Key to these interactions are signals transmitted between integrins and the IGF-I receptor (IGF-IR) when either or both are bound to their cognate ligands and we will review the current state of knowledge in this area. The IGF axis comprises many molecular components and we will also discuss the potential role of these species in cross-talk with the integrin receptor. With respect to integrin ligands, we will mainly focus on the well-characterized interactions of the two extracellular matrix (ECM) glycoproteins fibronectin (FN) and vitronectin (VN) with cell-surface ligands, and, how this affects activity through the IGF axis. However, we will also highlight the importance of other integrin activation mechanisms and their impact on IGF activity.
Collapse
Affiliation(s)
- James Beattie
- Department of Oral Biology, Leeds Dental Institute, University of Leeds, Leeds, UK.
| | | | | |
Collapse
|
41
|
Synthetic peptide-acrylate surfaces for long-term self-renewal and cardiomyocyte differentiation of human embryonic stem cells. Nat Biotechnol 2010; 28:606-10. [PMID: 20512120 DOI: 10.1038/nbt.1629] [Citation(s) in RCA: 325] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Accepted: 04/01/2010] [Indexed: 11/08/2022]
Abstract
Human embryonic stem cells (hESCs) have two properties of interest for the development of cell therapies: self-renewal and the potential to differentiate into all major lineages of somatic cells in the human body. Widespread clinical application of hESC-derived cells will require culture methods that are low-cost, robust, scalable and use chemically defined raw materials. Here we describe synthetic peptide-acrylate surfaces (PAS) that support self-renewal of hESCs in chemically defined, xeno-free medium. H1 and H7 hESCs were successfully maintained on PAS for over ten passages. Cell morphology and phenotypic marker expression were similar for cells cultured on PAS or Matrigel. Cells on PAS retained normal karyotype and pluripotency and were able to differentiate to functional cardiomyocytes on PAS. Finally, PAS were scaled up to large culture-vessel formats. Synthetic, xeno-free, scalable surfaces that support the self-renewal and differentiation of hESCs will be useful for both research purposes and development of cell therapies.
Collapse
|
42
|
Hajj GNM, Santos TG, Cook ZSP, Martins VR. Developmental expression of prion protein and its ligands stress-inducible protein 1 and vitronectin. J Comp Neurol 2010; 517:371-84. [PMID: 19760599 DOI: 10.1002/cne.22157] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Prion protein (PrP(C)) is the normal isoform of PrP(Sc), a protein involved in neurodegenerative disorders. PrP(C) participates in neuritogenesis, neuroprotection, and memory consolidation through its interaction with the secreted protein stress-inducible protein 1 (STI1) and the extracellular matrix protein vitronectin (Vn). Although PrP(C) mRNA expression has been documented during embryogenesis, its protein expression patterns have not been evaluated. Furthermore, little is known about either Vn or STI protein expression. In this study, PrP(C), STI1, and Vn protein expression was explored throughout mouse embryonic life. We found that the distributions of the three proteins were spatiotemporally related. STI1 and Vn expression became evident at E8, earlier than PrP(C), in the nervous system and heart. At E10, we observed, in the spinal cord, a gradient of expression of the three proteins, more abundant in the notochord and floor plate, suggesting that they can have a role in axonal growth. As development proceeded, the three proteins were detected in other organs, suggesting that they may play a role in the development of nonneural tissues as well. Finally, although STI1 and Vn are PrP(C) ligands, their expression was not altered in PrP(C)-null mice.
Collapse
Affiliation(s)
- Glaucia N M Hajj
- Ludwig Institute for Cancer Research, São Paulo 01323-903, Brazil
| | | | | | | |
Collapse
|
43
|
Bronson R. What the sperm says and the egg hears - a tale of two proteins and more. Am J Reprod Immunol 2009; 62:357-64. [PMID: 19895373 DOI: 10.1111/j.1600-0897.2009.00758.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
While considerable information exists regarding the early interactions of spermatozoon and egg that lead to successful fertilization, the molecular biology of events that result in the incorporation of the spermatozoon within the cortical ooplasm is largely undefined. There is circumstantial evidence suggesting that this process involves the interactions of specific oolemmal receptors and their ligands on sperm that bear similarities to mechanisms used in phagocytosis by macrophages. We have postulated that the egg may act as a 'non-professional phagocyte' during its association with the spermatozoon. This review surveys those events, provides an historical context, and creates a paradigm for further investigation.
Collapse
Affiliation(s)
- Richard Bronson
- Departments of Obstetrics & Gynecology and Pathology, Stony Brook University Medical Center, Stony Brook, NY 11794-8091, USA.
| |
Collapse
|
44
|
Adhikari TB, Bai J, Meinhardt SW, Gurung S, Myrfield M, Patel J, Ali S, Gudmestad NC, Rasmussen JB. Tsn1-mediated host responses to ToxA from Pyrenophora tritici-repentis. MOLECULAR PLANT-MICROBE INTERACTIONS : MPMI 2009; 22:1056-1068. [PMID: 19656041 DOI: 10.1094/mpmi-22-9-1056] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The toxin sensitivity gene Tsn1 interacts with Ptr ToxA (ToxA), a host-selective toxin produced by the necrotrophic fungus Pyrenophora tritici-repentis. The molecular mechanisms associated with cell death in sensitive wheat cultivars following ToxA application are not well understood. To address this question, we used the Affymetrix GeneChip Wheat Genome Array to compare gene expression in a sensitive wheat cultivar possessing the Tsn1 gene with the insensitive wheat cv. Nec103, which lacks the Tsn1 gene. This analysis was performed at early timepoints after infiltration with ToxA (e.g., 0.5 to 12 h postinfiltration [hpi]); at this time, ToxA is known to internalize into mesophyll cells without visible cell death symptoms. Gene expression also was monitored at later timepoints (24 to 48 hpi), when ToxA causes extensive damage in cellular compartments and visible cell death. At both early and late timepoints, numerous defense-related genes were induced (2- to 197-fold increases) and included genes involved in the phenylpropanoid pathway, lignification, and the production of reactive oxygen species (ROS). Furthermore, a subset of host genes functioning in signal transduction, metabolism, and as transcription factors was induced as a consequence of the Tsn1-ToxA interaction. Nine genes known to be involved in the host defense response and signaling pathways were selected for analysis by quantitative real-time polymerase chain reaction, and the expression profiles of these genes confirmed the results obtained in microarray experiments. Histochemical analyses of a sensitive wheat cultivar showed that H(2)O(2) was present in leaves undergoing cell death, indicating that ROS signaling is a major event involved in ToxA-mediated cell death. The results suggest that recognition of ToxA via Tsn1 triggers transcriptional reprogramming events similar to those reported for avirulence-resistance gene interactions, and that host-derived genes play an important role in the modulation of susceptibility to P. tritici-repentis.
Collapse
Affiliation(s)
- Tika B Adhikari
- Department of Plant Pathology, North Dakota State University, Fargo, ND, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Røger M, Høgåsen K, Solum NO, Mollnes TE, Hovig T. Vitronectin Inhibits Blood Platelet Aggregation. Platelets 2009; 4:225-9. [DOI: 10.3109/09537109309013222] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
46
|
The correlation between the adsorption of adhesive proteins and cell behaviour on hydroxyl-methyl mixed self-assembled monolayers. Biomaterials 2009; 30:307-16. [DOI: 10.1016/j.biomaterials.2008.09.048] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Accepted: 09/23/2008] [Indexed: 12/24/2022]
|
47
|
Meyers SR, Khoo X, Huang X, Walsh EB, Grinstaff MW, Kenan DJ. The development of peptide-based interfacial biomaterials for generating biological functionality on the surface of bioinert materials. Biomaterials 2008; 30:277-86. [PMID: 18929406 DOI: 10.1016/j.biomaterials.2008.08.042] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Accepted: 08/28/2008] [Indexed: 01/08/2023]
Abstract
Biomaterials used in implants have traditionally been selected based on their mechanical properties, chemical stability, and biocompatibility. However, the durability and clinical efficacy of implantable biomedical devices remain limited in part due to the absence of appropriate biological interactions at the implant interface and the lack of integration into adjacent tissues. Herein, we describe a robust peptide-based coating technology capable of modifying the surface of existing biomaterials and medical devices through the non-covalent binding of modular biofunctional peptides. These peptides contain at least one material binding sequence and at least one biologically active sequence and thus are termed, "Interfacial Biomaterials" (IFBMs). IFBMs can simultaneously bind the biomaterial surface while endowing it with desired biological functionalities at the interface between the material and biological realms. We demonstrate the capabilities of model IFBMs to convert native polystyrene, a bioinert surface, into a bioactive surface that can support a range of cell activities. We further distinguish between simple cell attachment with insufficient integrin interactions, which in some cases can adversely impact downstream biology, versus biologically appropriate adhesion, cell spreading, and cell survival mediated by IFBMs. Moreover, we show that we can use the coating technology to create spatially resolved patterns of fluorophores and cells on substrates and that these patterns retain their borders in culture.
Collapse
Affiliation(s)
- Steven R Meyers
- Department of Biomedical Engineering, Boston University, 44 Cummington Street, Boston, MA 02215, USA
| | | | | | | | | | | |
Collapse
|
48
|
Self-assembled monolayer films of phosphonates for bonding RGD to titanium. Clin Orthop Relat Res 2008; 466:977-84. [PMID: 18224380 PMCID: PMC2504684 DOI: 10.1007/s11999-008-0117-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2007] [Accepted: 01/04/2008] [Indexed: 01/31/2023]
Abstract
Modification of the implant surface with the Arg-Gly-Asp tripeptide (RGD) putatively facilitates osteoblast attachment for improved implant fixation in the laboratory. We compared the histomorphometric and mechanical performance of titanium implants coated with RGD using a novel interface of self-assembled monolayers of phosphonates (RGD/SAMP) and implants coated with RGD using the more conventional thiolate-gold interface (RGD/thiolate-gold). We hypothesized RGD/SAMP-coated implants would show greater bone ongrowth and implant fixation than RGD/thiolate-gold-coated ones. We implanted an RGD/SAMP-coated implant in one femur and an RGD/thiolate-gold-coated in the contralateral femur of 60 rats. At 2, 4, and 8 weeks after implantation, 10 rats were sacrificed for histologic evaluation and another 10 for biomechanical testing. Bone-implant ongrowth and implant force-to-failure of the two implants were similar at all times. Although RGD/SAMP-coated implants did not show superior bone ongrowth and implant fixation, RGD/SAMP-coated implants have at least equally good histomorphometric and mechanical in vivo performance as RGD/thiolate-gold-coated ones. Additional in vivo characterization of self-assembled monolayer films of phosphonates as interface to bond RGD to titanium is needed to explore its full potential and seems justified based on the results of this study.
Collapse
|
49
|
Secchi AG, Grigoriou V, Shapiro IM, Cavalcanti-Adam EA, Composto RJ, Ducheyne P, Adams CS. RGDS peptides immobilized on titanium alloy stimulate bone cell attachment, differentiation and confer resistance to apoptosis. J Biomed Mater Res A 2008; 83:577-84. [PMID: 17503524 DOI: 10.1002/jbm.a.31007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A major cause of implant failure in skeletal tissues is failure of osseointegration, often due to lack of adhesion of cells to the titanium (Ti) alloy interface. Since arginine-glycine-aspartic acid (RGD)-containing peptides have been shown to regulate osteoblast adhesion, we tested the hypothesis that, bound to a Ti surface, these peptides would promote osteoblasts differentiation, while at the same time inhibit apoptosis. RGDS and RGES (control) peptides were covalently linked to Ti discs using an APTS linker. While the grafting of both RGDS and RGES significantly increased Ti surface roughness, contact angle analysis showed that APTS significantly increased the surface hydrophobicity; when the peptides were tethered to Ti, this was reduced. To evaluate attachment, MC3T3-E1 osteoblast cells were grown on these discs. Significantly more cells attached to the Ti-grafted RGDS then the Ti-grafted RGES control. Furthermore, expression of the osteoblasts phenotype was significantly enhanced on the Ti-grafted RGDS surface. When cells attached to the Ti-grafted RGDS were challenged with staurosporine, an apoptogen, there was significant inhibition of apoptosis; in contrast, osteoblasts adherent to the Ti-grafted RGES were killed. It is concluded that RGD-containing peptides covalently bonded to Ti promotes osteoblasts attachment and survival with minimal changes to the surface of the alloy. Therefore, such modifications to Ti would have the potential to promote osseointegration in vivo.
Collapse
Affiliation(s)
- A G Secchi
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Marioli DJ, Zarkadis IK. The vitronectin gene in rainbow trout: cloning, expression and phylogenetic analysis. FISH & SHELLFISH IMMUNOLOGY 2008; 24:18-25. [PMID: 17981477 DOI: 10.1016/j.fsi.2007.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2007] [Revised: 06/19/2007] [Accepted: 07/01/2007] [Indexed: 05/25/2023]
Abstract
Vitronectin is a major cell adhesion glycoprotein that is found in plasma and the extracellular matrix. Vitronectin consists of an N-terminal somatomedin B domain and two hemopexin-like domains and controls functions including cell adhesion, migration, haemostasis and immune defence. In order to study the molecular evolution of the complement lytic pathway regulation, we have cloned and characterized the vitronectin gene from rainbow trout (Oncorhynchus mykiss). The deduced amino acid sequence of trout vitronectin exhibits 45%, 46%, 47% and 63% identity with human, chicken, Xenopus and zebrafish orthologs, respectively. The domain architecture of the trout vitronectin, consisting of a somatomedin B domain and two hemopexin-like domains, resembles that of mammalian vitronectins. Analysis of partial genomic clones shows that trout vitronectin gene exhibits the same exon-intron organization profile as the human ortholog gene. The trout vitronectin gene is probably present as a single copy in the trout genome, showing a differential expression pattern among tissues investigated.
Collapse
Affiliation(s)
- Dimitra J Marioli
- Department of Biology, School of Medicine, University of Patras, Rion 26500, Patras, Greece
| | | |
Collapse
|